ArticlePDF Available

Pathogenesis and prevention of biomaterial centered infection

Authors:

Abstract

One of the major drawbacks in the use of biomedical materials is the occurrence of biomaterials centered infections. After implantation, the host interacts with a biomaterial by forming a conditioning film on its surface and an immune reaction towards the foreign material. When microorganisms can reach the biomaterials surface they can adhere to it. Adhesion of microorganisms to an implant is mediated by their physico-chemical surface properties and the properties of the biomaterials surface itself. Subsequent surface growth of the microorganisms will lead to a mature biofilm and infection, which is difficult to eradicate by antibiotics. The purpose of this review is to give an overview of the mechanisms involved in biomaterials centered infection and the possible methods to prevent these infections.
JOURNAL OF MATERIALS SCIENCE: MATERIALS IN MEDICINE 13 (2002) 717±722
Pathogenesis and prevention of biomaterial
centered infections
B. GOTTENBOS, H. J. BUSSCHER*, H. C. VAN DER MEI
Department of Biomedical Engineering, University of Groningen, Faculty of Medical
Sciences, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
P. NIEUWENHUIS
Department of Histology and Cell Biology, University of Groningen, Faculty of Medical
Sciences, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
E-mail: h.j.busscher@med.rug.nl
One of the major drawbacks in the use of biomedical materials is the occurrence of
biomaterials centered infections. After implantation, the host interacts with a biomaterial by
forming a conditioning ®lm on its surface and an immune reaction towards the foreign
material. When microorganisms can reach the biomaterials surface they can adhere to it.
Adhesion of microorganisms to an implant is mediated by their physico-chemical surface
properties and the properties of the biomaterials surface itself. Subsequent surface growth of
the microorganisms will lead to a mature bio®lm and infection, which is dif®cult to eradicate
by antibiotics. The purpose of this review is to give an overview of the mechanisms involved
in biomaterials centered infection and the possible methods to prevent these infections.
#2002 Kluwer Academic Publishers
1. Introduction
Foreign materials are used more and more in modern
medicine after trauma, oncological surgery or wear to
replace, support or restore human body function, for
example in hip prostheses, prosthetic heart valves or
catheters. The extended use of these materials, usually
referred to as biomaterials, has some major drawbacks.
One of these is the possible occurrence of biomaterials
centered infections (BCI) [1]. The incidence of this type
of infections varies from 4% for hip prostheses [2] to
100% for urinary tract catheters after 3 weeks use [3] (see
Table I). In BCI microorganisms are present in close
association with the biomaterials surface forming a so-
called bio®lm. BCI can cause severe problems, from
disfunctioning of the implanted device to lethal sepsis of
the patient. Furthermore, treatment of BCI is compli-
cated, as microorganisms in a bio®lm are more resistant
to antibiotics [4] than their planktonic counterparts [5].
As a consequence and if possible, the only remedy is
removal of the infected implant at the expense of
considerable costs and patient's suffering. A more
convenient way to deal with this problem is to prevent
the development of an infectious bio®lm on the
biomaterials surface. To achieve this, a thorough under-
standing of how these bio®lms develop is necessary.
2. Host±biomaterials interactions
2.1. Conditioning ®lms
The interaction of biomaterials with the body of the host
depends on the place where the implant or device is
situated, for example in the oropharyngeal cavity, the
urinary tract, different body tissues or in the circulatory
system. When a biomaterial is inserted, ®rst a so-called
conditioning ®lm from organic matter present in the
surrounding ¯uid is deposited on the biomaterials
surface. Depending on the body site the surrounding
¯uid can be saliva, urine, tear ¯uid, tissue ¯uid, serum, or
blood, and the conditioning ®lm will mostly consist of
adsorbed proteins, which for serum are mainly albumin,
immunoglobulin, ®brinogen, and ®bronectin. The
composition of the conditioning ®lm depends on the
physico-chemical properties, i.e. chemical composition,
hydrophobicity and charge, of the biomaterials surface.
For example, on polyethylene hydrophobicity gradients
exposed to blood serum it was found that at the
hydrophobic end less proteins were adsorbed with
relatively more ®brinogen, while on the hydrophilic
end more albumin was present [6]. In time, smaller
proteins like albumin are usually replaced by higher
molecular weight proteins, like ®brinogen and ®bro-
nectin. With blood contacting biomaterials, also blood
cells will adhere to the biomaterials surface. Especially
adhesion of blood platelets to arti®cial vascular grafts
can initiate the blood coagulase cascade, causing
thrombosis, a frequent complication in these applica-
tions. Another unwanted phenomenon at the biomaterials
surface is calci®cation of the implant, which can
decrease the necessary ¯exibility of, for example,
prosthetic heart valves. Ideally host derived cells will
colonize the implanted biomaterials, forming a thin
capsule around the implant.
*Author to whom correspondence should be addressed.
0957±4530 #2002 Kluwer Academic Publishers Review Paper 717
2.2. Immune reactions
The host will also actively interact with the biomaterials
surface if it is invasively implanted, as this is a normal
reaction to any foreign body that enters the host. The
coagulation cascade and complement system are acti-
vated, leading to formation of a ®brin network and
opsonization of the biomaterial [7, 8]. These processes
will attract and activate the innate immune system, i.e.
macrophages and polymorphonuclear cells, leading to
in¯ammation [9, 10]. This immune response can dis-
appear when the wound is healed and the biomaterial is
encapsulated. However, in many cases the host±
biomaterials interface remains in a state of chronic
in¯ammation, as few metals and plastics are chemically
inert in the warm, wet, oxygenated environment of living
tissues, causing the release of in¯ammatory compounds
from the biomaterial, like corrosion products, plastici-
zers, and monomers [1, 11]. Chronic in¯ammation
impairs host cell growth on the implant [12] and can
cause chronic pain.
3. Pathogenesis of biomaterials centered
infections
The presence of a foreign material signi®cantly
compromises the host to cope with microorganisms. In
a classical study in man it was shown that the presence of
a subcutaneous suture reduced the required inoculum to
produce infection with Staphylococcus aureus,an
infamous virulent pathogen, from 10
6
to only 200
bacteria [13]. Furthermore, the relatively avirulent
Staphylococcus epidermidis, normally not capable of
establishing infection, is the most common infecting
organism in BCI [14].
3.1. Inoculation
Probably the most important factor determining the
occurrence of BCI is the chance that microorganisms will
reach the biomaterials surface. Biomaterials in contact
with the outer part of the body, for example intravenous
catheters, peritoneal dialysis catheters, or urinary tract
catheters are readily reached by microorganisms and
consequently have a higher incidence of BCI than fully
implanted biomaterials (0.5±100% vs. 0.1±7%) [2].
Microorganisms can reach a biomaterials implant in
several ways at several time points, which determines the
properties of the biomaterials surface they will meet.
Airborne microorganisms, which can be present in the
operating theater, can reach the surface as early as before
the implantation [15, 16] and interact with a bare
substratum surface, not even covered with a conditioning
®lm. Also during insertion of the biomaterial, micro-
organisms from the skin can be pushed towards the
implant surface. Furthermore, microorganisms from the
skin can contaminate the operation wound and reach the
implant surface through diffusion, active movement or
hematogenous transport. Perioperative contamination is
believed to be the most common cause of BCI [17].
Generally, it is also assumed that microorganisms can
reach the implant via the hematogenous route at any time
after implantation, causing so-called hematogenous
infections. As shown in Table II skin infections, surgical
or dental interventions, pneumonia, abscesses, or
bacteriuria can cause temporal or chronic bacteremia
resulting in infections [18]. In addition, microorganisms
can translocate from the gastro-intestinal tract to other
body parts [19]. When the microorganisms survive in the
bloodstream, they can be transported to the biomaterials
surface, establishing an infection. In this respect it is
especially interesting to note that it has been proposed
that macrophages play a role in transporting microorgan-
isms to the biomaterials surface [20] as some strains are
capable to survive within macrophages [21]. As the
biomaterials surface elicits a foreign body reaction in the
®rst weeks after implantation, and in the case of chronic
in¯ammation also hereafter, macrophages are speci®-
cally attracted to the biomaterials surface thus potentially
transporting microorganisms to the biomaterials implant
or device [22]. As hematogenous infection can happen
anytime, biomaterials implants are sometimes called
``microbial time bombs''.
The etiology of BCI can provide information about the
origin of the infecting organisms. Table III shows the
organisms found in examples of studies with vascular
grafts, hip and knee arthroplasties. S. epidermidis and S.
aureus, primarily skin inhabitants, are the predominant
infecting organisms [14], followed by Gram-negative
bacilli like Escherichia coli and Pseudomonas aerugi-
nosa, primarily present in the gastro-intestinal and
urinary tract, streptococci from the mouth and pneumo-
cocci from the respiratory tract [18, 23]. Interestingly,
except in the case of airborne microorganisms, the
T A B L E I I Distant infectious foci of hematogeneously infected hip
[17] and knee arthroplasties [18]
Distant foci Hip n27(%) Knee n72(%)
Cutaneous region 19 39
Urinary tract 15 19
Respiratory tract 15 14
Oral cavity 30 7
Gastrointestinal tract 4 6
Septic arthritis 0 3
Abdominal abscess 0 1
Unknown 19 11
T A B L E I Incidences of infection of different biomedical implants
and devices adapted from Dankert et al. [2] and arranged according to
body site
Body site Implant or device Incidence (%)
Urinary tract UT catheters 10±20
Percutaneous CV catheters 4±12
Temporary pacemaker 4
Short indwelling catheters 0.5±3
Peritoneal dialysis catheters 3±5
Subcutaneous Cardiac pacemaker 1
Soft tissue Mammary prosthesis 1±7
Intraocular lenses 0.13
Circulatory system Prosthetic heart valve 1.88
Multiple heart valve 3.6
Vascular graft 1.5
Arti®cial heart* 40
Bones Prosthetic hip 2.6±4.0
Total knee 3.5±4
*From experiments in calves and sheep.
718
infecting organisms usually originate from the hosts
micro¯ora. There is evidence that the host might be
immuno-tolerant to some of these microorganisms,
which has been especially investigated for the intestinal
micro¯ora [24±26]. This might be an overlooked
virulence factor for causing BCI. As immuno-tolerated
microorganisms can survive longer in the blood without
being attacked by the hosts immune system, they have a
bigger chance to reach an implanted biomaterial.
3.2. Microbial adhesion
When microorganisms have reached the biomaterials
surface, initial microbial adhesion can occur. Microbial
adhesion is mediated by speci®c interactions between
cell surface structures and speci®c molecular groups on
the substratum surface [14], or when viewed from an
overall, physico-chemical view-point by non-speci®c
interaction forces, including Lifshitz±Van der Waals
forces, electrostatic forces, acid±base interactions, and
Brownian motion forces [27]. Speci®c interactions are in
fact non-speci®c forces acting on highly localized
regions of the interacting surfaces over distances smaller
than 5 nm, while non-speci®c interaction forces have a
long-range character and originate from the entire body
of the interacting surfaces, as is shown in Fig. 1 [28].
Upon approach of a surface, organisms will be attracted
or repelled by the surface, depending on the resultant of
the different non-speci®c interaction forces. Thus the
physico-chemical surface properties of the biomaterial,
with or without conditioning ®lm or epithelial cells, and
microorganisms play a major role in this process.
The conditioning ®lm on the biomaterials surface (and
on the bacterial cell surface) plays an important role, as it
changes the physico-chemical properties of the inter-
acting surfaces. Most proteins are capable of reducing the
adhesion of microorganisms. Albumin is a strong
adhesion inhibitor, for unknown reasons, although
changes in hydrophobicity and sterical hindrance are
proposed mechanisms [14]. Fibronectin and ®brinogen
have been shown to promote the adhesion of S. aureus
and certain S. epidermidis strains, which is mediated by
speci®c adhesive cell structures directed to these proteins
[14, 29].
The adhesion mechanism of late hematogenously
transported microorganisms is unclear, as by that time
the biomaterial will be covered with host derived cells,
which will decrease the adhesion probability of
microorganisms [1]. Hematogenous infections are,
besides with intravascular prostheses, almost exclusively
reported with orthopedic implants [17, 18]. Orthopedic
implants are usually not completely integrated within
host tissue, as they consist often of metal parts, which are
not easily colonized by host tissue cells. The uncovered
metal surface can be colonized readily by microorgan-
isms. Another explanation could be that the repeated
hinging of orthopedic implants, for example in knee
prostheses, can cause cell damage, providing adhesive
sites for microorganisms [1].
When the microorganisms have adhered to a bioma-
terials surface they are protected against phagocytosis, as
the microorganism and biomaterial together are too large
to ingest. Furthermore Zimmerli and coworkers reported
that the activity of phagocytes and polymorphonuclear
leukocytes is decreased in the presence of a biomaterial
[30, 31]. After adhesion to biomaterials most micro-
organisms start secreting slime and embed themselves in
a slime layer, the glycocalix, which is an important
virulence factor for BCI and which explains the
extraordinary prevalence of slime producing S. epider-
midis in BCI [14]. The glycocalix provides protection
against humoral and excreted cellular immune compo-
nents, as these cannot readily diffuse through the slime
layer [4] and once a glycocalix has formed a BCI with all
its complications, including ultimately removal of the
implant, seems almost inevitable. However, to do real
damage the adhering microorganisms ®rst have to grow.
T A B L E I I I Microbial etiology of vascular graft [23] and hematogenous orthopedic implant [17, 18] infections
Germ Vascular grafts (%) Orthopedic implants (%)
Abdominal n17Inguinal n60Popliteal n8Hip n27Knee n72
S. aureus 14 40 33 52 52
S. epidermidis 7131744
Streptococci 14 8 25 22 14
Pneumococci n.d. n.d. n.d. 7 6
E. coli 42 9 0 7 11
Proteus species 3 11 0 4 4
other Gr. neg. bacilli 0 8 17 4 4
Other bacteria 10 5 0 0 1
Candida species 3 1 0 0 0
Unknown 7 5 8 0 0
n.d. not determined. Values for nindicate number of patients.
Figure 1 At separation distances of 450 nm, only attractive Van der
Waals forces occur. At 10±20nm, Van der Waals and repulsive
electrostatic interactions in¯uence adhesion. At 55 nm, short-range
interactions can occur, irreversibly binding a bacterium to a surface.
Adapted from Busscher and Weerkamp [27].
719
3.3. Microbial growth
Initial microbial growth, i.e. the growth in the ®rst hours
after microbial adhesion, is another important factor for
the outcome of BCI, as bio®lm organisms become more
resistant to the immune system while the bio®lm
matures. Growth of sessile microorganisms has been
mainly studied in vitro [32]. Barton et al. [33] found that
growth of sessile P. aeruginosa, S. epidermidis and E.
coli depended on the biomaterial involved. Only for P.
aeruginosa they could ®nd a correlation between
physico-chemical properties of the biomaterials surface
and initial growth rates. Also proteins in the conditioning
®lm can in¯uence the growth rates of adhering
microorganisms. Poelstra et al. found that growth rates
of P. aeruginosa decreased in the presence of pooled
immunoglobulin G [34].
Van Loosdrecht et al. [35] concluded that adhesion of
bacteria does not directly in¯uence their metabolism and
growth yield. Changes in growth rate due to adhesion of
bacteria were suggested to be mainly the result of
changes in nutrient availability. Depending on the
amount of adsorbed nutrients and whether adsorption is
easily reversed, growth rates of adhering bacteria can be
decreased or increased with respect to the growth of their
planktonic counterparts. Another mechanism, causing a
decrease in the growth rates of E. coli after adhesion was
proposed to be the strong attraction to positively charged
biomaterials surfaces [36].
There is little information about microbial growth in
vivo. Barth et al. followed the number of bacteria for 48 h
on subcutaneously implanted polymeric and metal
biomaterials in rabbits, after inoculation at the time of
implantation [37]. They found, as is shown in Fig. 2, that
the number of non-slime producing S. epidermidis
decreased directly after implantation, probably due to
action of the host immune system, while the number of
slime producing S. epidermidis and S. aureus increased
in the ®rst 8±12 h to a maximum, whereafter their
number decreased again. Here also materials differences
played a role. S. aureus grew faster on the metal, while S.
epidermidis grew faster on polymeric biomaterials. An
important difference between in vitro and in vivo
experiments is the presence of the immune system.
Although unprotected bacteria are eradicated by the
immune system, it has been reported that those
microorganisms that do survive accelerate their growth
rates under in¯uence of cytokines excreted by macro-
phages [38].
3.4. Consequences of BCI
The presence of a microbial bio®lm on biomaterials
impairs the function of the implant or device and/or
worsens the clinical state of the patient. Examples with
non-implanted devices are voice prostheses, which are
situated between the trachea and the upper digestive
tract, as is shown in Fig. 3, or urinary tract catheters. The
action of the voice prosthesis is impaired by bio®lm
formation, because microorganisms block the valve
mechanism, or cause leakage of food into the trachea
[39]. As a consequence the prosthesis has to be replaced
every 4 months on average [40]. Urinary tract catheters
rarely escape colonization by microorganisms causing
blockage or, more seriously, bacteriuria [41]. Infections
of indwelling catheters, like for example central venous
catheters, often results in bacteremia which can cause
sepsis and endocarditis. Totally implanted prostheses
have lower rates of infections, but the consequences are
often more serious. Especially infections of implants in
the circulatory system, i.e. prosthetic valves and vascular
grafts, yield a high mortality (70% and 50% respectively
[42]). Infection of deep tissue implants, for example
Figure 2 The numbers of colony forming units per disk of (a) PMMA,
(b) UHMW-PE and (c) titanium vanadium aluminum alloy after
different in vivo implantation times. SE-360 (black) is a slime
producing and SP-2 (gray) is a non-slime producing strain of S.
epidermidis. White bars represent S. aureus. Note the decline in
adhering non-slime producing S. epidermidis in time. Adapted from
Barth et al. [36].
Figure 3 The voice prosthesis (a) and anatomy after total laryngectomy
(b). The voice prosthesis is placed in the tracheo-esophageal shunt, an
area which is heavily inhabited by microorganisms.
720
orthopedic implants or mammary prostheses, will usually
result in less serious complications like pain, swelling
and loosening of the implant, although mortalities up to
20% are reported with orthopedic implants [18, 43, 44].
The clinical signs of deep implant infections are reported
to appear up to a year after microbial seeding [45].
Apparently the infectious bio®lm can stay silent for a
long period, and probably a signi®cant part of the
infections is never recognized. Recent investigations at
our laboratory revealed that the so-called aseptic
loosening, i.e. loosening of an orthopedic implant
without microorganisms present, is often diagnosed
falsely as aseptic. As standard microbiological techni-
ques are used to test for the presence of infectious
microorganisms, slow growing bio®lm organisms often
remain undetected. Similarly, it has been reported that
®ve of 28 removed scleral explants were covered with a
bio®lm, while clinical signs of infection were only
present in one out of these ®ve cases [46]. Thus the
incidence of problems associated with BCI is possibly
higher than generally assumed.
4. Treatment and prevention of BCI
4.1. Treatment
Treatment of an established BCI is dif®cult, as the
minimal inhibitory concentration (MIC) of antimicrobial
agents, necessary to kill the microorganisms, is sig-
ni®cantly higher for microorganisms in a bio®lm than for
planktonic ones [4, 5]. As antibiotics have little effect on
BCI, the standard procedure for infected orthopedic
prostheses is the removal of the implant and implantation
of an antibiotic releasing device at the implant site. A
new prosthesis is inserted when the implant site is free of
microorganisms, usually six months later. For many
implants, especially those in the circulatory system,
removal of the implant is dangerous, and a high mortality
is associated with these infections. Much research has
been done to make bio®lm bacteria more susceptible to
antibiotics. Ultrasonic treatment of the infecting bio®lm,
for example, has been shown to enhance the action of
antibiotics towards these bio®lm bacteria [47]. Also
application of an electrical ®eld yields enhanced effects
of antibiotic treatment, the so-called bioelectric effect
[48]. The application of these techniques in patients
could facilitate the treatment of BCI with antibiotics in
the future.
4.2. Prevention
Surgeons take considerable effort in preventing the
contamination of implants with microorganisms during
implantation. Although application of prophylactic
antibiotics and better operation hygiene has reduced the
incidence of BCI the last four decades, still a signi®cant
number of patients suffer from these infections.
Different strategies seem useful to prevent BCI. In
general it is aimed to reduce the attractive force between
bacteria and biomaterials surface by optimizing the
physico-chemical surface properties of the biomaterial.
Bacterial adhesion is low, for example, on extremely
hydrophobic surfaces [49, 50], while also more nega-
tively charged biomaterials attract less bacteria [51].
Albumin and heparin coatings have shown to decrease
the adhesiveness of biomaterials [52].
However, microorganisms always seem to be able to
adhere to some extent to solid materials. Moreover, when
proteins are present they can cover an anti-adhesive
biomaterial, and be anchors for microorganisms to
adhere to. Another approach to prevent bio®lm formation
is to prevent the growth of adhering microorganisms.
This can be achieved by application of antimicrobial
agents near the biomaterials surface. One way to do this
is the design of antibiotic releasing biomaterials.
Examples are gentamicin-loaded bone cement and
silver-loaded catheters [53, 54]. A disadvantage of
these applications is that they usually only work for a
few days to weeks, as the amount of antibiotic that is
actually released is extremely limited and does not
exceed 15% of the total amount incorporated [55]. A
more dangerous problem with antibiotic releasing
materials and the low dose actually released is the
development of antibiotic resistant microbial strains [56].
A better approach would be to couple the antimicrobial
agent covalently onto the biomaterials surface, while
maintaining its activity. As in this approach the
antimicrobial agent can only reach the outside of the
microbial cells, it can only be employed with antibiotics
working at the level of the cell wall or membrane.
Polymers with incorporated quaternary ammonium
groups have shown such antimicrobial activity in vitro
[57, 58], thus these compounds might have the required
properties.
References
1. A. G. GRISTINA,Science 237 (1987) 1588.
2. J. DANKERT,A. H. HOGT and J. FEIJEN,CRC Crit. Rev.
Biocompat.2(1986) 219.
3. J. D. DENSTEDT,T. A. WOLLIN and G. REID,J. Endourol. 12
(1998) 493.
4. J. W. COSTERTON,P. S . ST EWAR T and E. P. GREENBERG,
Science 284 (1999) 1318.
5. P. GILBERT,J. DAS and I. FOLEY,Adv. Dent. Res. 11 (1997)
160.
6. H. T. SPIJKER,R. BOS,W. VAN OEVEREN,J. DE VRIES and
H. J. BUSSCHER,Coll. Surf. B: Biointerf. 15 (1999) 89.
7. L. TANG and J. W. EATON,Am. J. Clin. Pathol. 103 (1995) 466.
8. A. REMES and D. F. WILLIAMS,Biomaterials 13 (1992) 731.
9. T. E. MOLLNES,Vox Sang. 74 S2 (1998) 303.
10. J. M. ANDERSON,ASAIO Trans. 34 (1988) 101.
11. S. H. DOUGHERTY and R. L. SIMMONS,Curr. Probl. Surg. 19
(1982) 217.
12. J. H. JACKSON and C. G. COCHRANE,Hematol. Oncol. Clin.
North Am. 2(1988) 317.
13. S. D. ELEK and P. E . CON E N,Br. J. Exp. Pathol. 38 (1957) 573.
14. G. D. CHRISTENSEN,L. M. BADDOUR,D. L. HASTY,J. H.
LOW RA NC E and W. A. SIMPSON, in ``Infections Associated
with Indwelling Medical Devices'', edited by A. L. Bisno and
F. A. Waldvogel (American Society of Microbiology, Washington
DC, 1989) p. 27.
15. O. M. LIDWELL,E. J. LOWBURY,W. W H Y T E ,R. BLOWERS,
S. J. STANLEY and D. LOWE,Br. Med. J. Clin. Res. Ed. 285
(1982) 10.
16. J. CHARNLEY,Clin. Orthop. 87 (1972) 167.
17. A. AHLBERG,A. S. CARLSSON and L. LINDBERG,ibid. 137
(1978) 69.
18. S. BENGTSON,G. BLOMGREN,K. KNUTSON,A. WIGREN
and L. LIDGREN,Acta Orthop. Scand. 58 (1987) 529.
19. P. A. VAN-LEEUWEN,M. A. BOERMEESTER,A. P.
HOUDIJK,C. C. FERWERDA,M. A. CUESTA,S. MEYER
and R. I. WESDORP,Gut 35 (1994) S28.
721
20. E. M. MORA,M. A. CARDONA and R. L. SIMMONS,Arch.
Surg. 126 (1991) 157.
21. C. L. WELLS,M. A. MADDAUS and R. L. SIMMONS,ibid. 122
(1987) 48.
22. W. GU O,R. ANDERSSON,A. LJUNGH,X. D. WANG and S.
BENGMARK,Scand. J. Gastroenterol 28 (1993) 393.
23. G. PRINTZEN,Injury 27 S3 (1996) SC9.
24. R. DUCHMANN,E. SCHMITT,P. KNOLLE,B. K. MEYER-
ZUM and M. NEURATH,Eur. J. Immunol. 26 (1996) 934.
25. M. C. FOO and A. LEE,Infect. Immun. 6(1972) 525.
26. R. D. BERG and D. C. SAVAGE,ibid. 11 (1975) 320.
27. C. J. VAN OSS,Biofouling 4(1991) 25.
28. H. J. BUSSCHER and A. H. WEERKAMP,FEMS Microbiol.
Rev. 46 (1987) 165.
29. Y.H. AN and R. J. FRIEDMAN,J. Biomed. Mater. Res. 43 (1998)
338.
30. W. ZIMMERLI,P. D. LE W and F. A. WALDVOGEL,J. Clin.
Invest. 73 (1984) 1191.
31. W. ZIMMERLI,F. A. WALDVOGEL,P. VAUDAUX and U. E.
NYDEGGER,J. Infect. Dis. 146 (1982) 487.
32. G. G. GEESEY and D. C. WHITE,Annu. Rev. Microbiol. 44
(1990) 579.
33. A. J. BARTON,R. D. SAGERS and W. G . PI TT,J. Biomed.
Mater. Res. 32 (1996) 271.
34. K. A. POELSTRA,H. C. VAN DER MEI,B. GOTTENBOS,
D. W. GRAINGER,J. R. VAN HORN and H. J. BUSSCHER,
ibid. 50 (2000) 224.
35. M. C. M. VAN LOOSDRECHT,J. LYKLEMA,W. N OR DE and
A. J. B. ZEHNDER,Microbiol. Rev. 54 (1990) 75.
36. G. HARKES,J. DANKERT and J. FEIJEN,J. Biomater. Sci.
Polym. Ed. 3(1992) 403.
37. E. BARTH,Q. M. MYRVIK,W. WAG N E R and A. G.
GRISTINA,Biomaterials 10 (1989) 325.
38. P. B. VA N WACHE M,M. J. A. VAN LUYN,A. W. DE WIT,
D. RAATJES,M. HENDRIKS,M. L. P. M. VERHOEVEN and
L. CAHALAN,J. Biomed. Mater. Res. 35 (1997) 217.
39. H. F. MAHIEU,H. F. SAENE,H. J. ROSINGH and H. K.
SCHUTTE,Arch. Otolaryngol. 112 (1986) 321.
40. F. A. VAN DEN HOOGEN,M. J. OUDES,G. HOMBERGEN,
H. F. NIJDAM and J. J. MANNI,ibid. 116 (1996) 119.
41. J. C. NICKEL,J. W. COS TERTON,R. J. MCLEAN and M.
OLSON,J. Antimicrob. Chemother. Suppl. A 33 (1994) 31.
42. K. H. MAYER and S. C. SCHOENBAUM,Prog. Cardiovasc.
Dis. 25 (1982) 43.
43. G. HUNTER and D. DANDY,J. Bone Joint Surg. Br. 59 (1977)
293.
44. R. H. FITZGERALD and D. R. JONES,Am. J. Med. 78 (1985)
225.
45. G. MANILOFF,R. GREENWALD,R. LASKIN and C. SINGER,
Clin. Orthop. 223 (1987) 194.
46. R. H. ASARIA,J. A. DOWNIE,L. MCLAUGLIN-BORLACE,
N. MORLET,P. MU NR O and D. G. CHARTERIS,Retina 19
(1999) 447.
47. A. M. REDISKE,B. L. ROEDER,M. K. BROWN,J. L.
NELSON,R. L. ROBISON,D. O. DRAPER,G. B.
SCHAALJE,R. A. ROBISON and W. G. P IT T,Antimicrob.
Agents Chemother. 43 (1999) 1211.
48. J. W. COSTERTON,B. ELLIS,K. LAM,F. JOHNSON and A. E.
KHOURY
,ibid. 38 (1994) 2803.
49. E. P. EVERAERT,H. F. MAHIEU,B. VAN DE BELT-
GRITTER,A. J. PEETERS,G. J. VERKERKE,H. C. VAN
DER MEI and H. J. BUSSCHER,Arch. Otolaryngol. Head Neck
Surg. 125 (1999) 1329.
50. J. TSIBOUKLIS,M. STONE,A. A. THORPE,P. GRAHAM,V.
PETERS,R. HEERLIEN,J. R. SMITH,K. L. GREEN and T. G .
NEVELL,Biomaterials 20 (1999) 1229.
51. A. H. HOGT,J. DANKERT and J. FEIJEN,J. Biomed. Mater.
Res. 20 (1986) 533.
52. J. R. KEOGH and J. W. EATON,J. Laborat. Clin. Med. 124
(1994) 537.
53. A. S. CARLSSON,G. JOSEFSSON and L. LINDBERG,J. Bone
Joint Surg. 60 (1978) 1059.
54. H. AKIYAMA and S. OKAMOTO,J. Urol. 121 (1979) 40.
55. H. VAN DE BELT,D. NEUT,D. R. A. UGES,W. SCHENK,J. R.
VAN H OR N,H. C. VAN DER MEI and H. J. BUSSCHER,
Biomaterials 21 (2000) 1981.
56. H. VAN DE BELT,D. NEUT,J. VAN HORN,H. C. VAN DER
MEI,W. SCHENK and H. J. BUSSCHER,Nat. Med. 5(1999)
358.
57. R. G. FLEMMING,C. C. CAPELLI,S. L. COOPER and R. A.
PROCTOR,Biomaterials 21 (2000) 273.
58. R. EL-KENAWY,F. I. ABDEL-HAY,A. EL-RAHEEM,R. EL-
SHANSHOURYand M. H. EL-NEWEHY,J. controled Release 50
(1998) 145.
Received 11 January 2001
and accepted 7 February 2002
722
... In comparison, the untreated sample (control) had no apparent effect on the inhibition of bacterial colonization or killing of attached bacteria. The antibacterial activity of zirconia-based materials after laser treatment can be evaluated based on the following three factors [68]: (1) physicochemical (surface wettability), (2) chemical (oxide layer composition, thickness, and charge-carrier characteristics), and physical changes (surface roughness and topography). Bacteria move or are transferred to the surface of a material through the effects of physical forces, such as Brownian motion, van der Waals attraction forces, gravitational forces, surface electrostatic charges, and hydrophobic interactions [63,68]. ...
... The antibacterial activity of zirconia-based materials after laser treatment can be evaluated based on the following three factors [68]: (1) physicochemical (surface wettability), (2) chemical (oxide layer composition, thickness, and charge-carrier characteristics), and physical changes (surface roughness and topography). Bacteria move or are transferred to the surface of a material through the effects of physical forces, such as Brownian motion, van der Waals attraction forces, gravitational forces, surface electrostatic charges, and hydrophobic interactions [63,68]. There are two generally accepted theories on the adhesion of bacteria to solid surfaces. ...
Article
3 mol% yttria-stabilized zirconia ceramics have been gaining attention as promising restorative materials that are extensively used in dental implant applications. However, implant failure due to bacterial infection and its bioinert surface slow osseointegration in vivo, which are significant issues in clinical applications. In this work, surface modification was achieved using a continuous wave carbon dioxide laser at a wavelength of 10.6 µm in an air atmosphere. Changes in the surface characteristics were evaluated using X-ray diffraction (XRD), field-emission scanning electron microscopy (FESEM), atomic force microscopy (AFM), and 2D roughness and hardness tests. The bioactivity of the laser-treated samples was studied by examining their behavior when immersed in the SBF solution. The formation of the hydroxyapatite phase on the laser-treated sample was much more uniform than that of its untreated counterparts. The antibacterial properties of surface-treated zirconia ceramics against Streptococcus mutans and Escherichia coli bacteria were rigorously examined. These results indicate that the laser-induced nanoscale grooves significantly improved antibacterial activity by creating hydrophobic surfaces. The cellular response was evaluated for 7 days on microtextures on the zirconia surfaces and an untreated sample with MC3T3-E1 pre-osteoblast cell line cultured under basal conditions. Surface topography was revealed to improve the cellular response with increased metabolic activity compared to the untreated sample and showed modulation of cell morphology for the entire time. These results suggest that laser modification can be an appropriate non-contact method for designing nanoscale microtextures to improve the biological response and antibacterial behavior of zirconia ceramics in restorative dentistry.
... flagella, capsule, and extracellular polymeric substances (EPS) [5][6][7]. The interactions between these appendages and the solid surfaces involve chemical and physical bonds such as weak hydrogen bonds, Van der Waals forces of attraction, and ligand-receptor interactions [5,6]. ...
... flagella, capsule, and extracellular polymeric substances (EPS) [5][6][7]. The interactions between these appendages and the solid surfaces involve chemical and physical bonds such as weak hydrogen bonds, Van der Waals forces of attraction, and ligand-receptor interactions [5,6]. These interactions also enhance the ability of the cells to acquire metals and compounds present in the surrounding environment and the microorganisms utilize these for their metabolic activities and growth. ...
Article
Exopolymeric substances (EPS) produced by bacterial cells play a crucial role in the interaction of the cells with the surrounding environment. Halobacillus trueperi manxer mangrove‐16, an adhered bacterial isolate from the mangrove ecosystem was found to produce EPS that was observed by Alcian blue staining and congo red‐coomassie blue agar. The EPS of the bacterial isolate exhibited emulsifying properties. Purification of the EPS by dialysis showed an emulsification index of 80% with hexadecane. Qualitative analysis and Fourier's Infrared spectroscopy (FTIR) revealed that the EPS was a glycoprotein in nature. The EPS showed no surface‐active properties. Further exploration of the potential of the EPS interaction with metal solutions showed the ability of the bioemulsifier to cause precipitation in the metal solutions and particularly change the color of the Chromium (VI) solution. The scanning electron microscopy‐energy‐dispersive X‐ray spectroscopy (SEM‐EDS) of the cells and EPS particularly indicated the interaction of the EPS with the (Fe0) zerovalent iron nanoparticles and its effect on the cells and EPS of the bacteria. It is therefore concluded that the EPS is a crucial component that anchors the bacteria to particulate matter in the mangrove ecosystem and also plays an important role in interaction with metals and hydrocarbons.
... the health of users [1][2][3]. Several medical devices that possess high risk of BCI include sutures, urinary catheters, IUDs, vascular grafts, and orthopedic devices [4]. ...
Article
Biomaterial-Centered Infection (BCI) is a significant issue in the implantation of medical devices, primarily caused by the formation of bacterial biofilms on the device surface. One potential solution to address this problem is the use of antibacterial coatings. This study examines the effectiveness of everolimus as an antibacterial coating agent with polydopamine (PDA) as an intermediate layer. The commonly used biomaterial for medical scaffolds, poly(lactic acid) (PLA), was coated with everolimus after being submerged in PDA for 24 hours. The coated PLA was then subjected to antibacterial analysis, including culturing Gram-positive Staphylococcus aureus and Gram-negative Escherichia coli bacteria in Luria-Bertani broth, bacterial count tests, and disc inhibition tests. The results showed that everolimus has antibacterial properties, but its efficacy varies with different types of bacteria adhering to the biomaterial surfaces. Specifically, the everolimus coating was found to be more effective in killing Gram-positive S. aureus. The absence of inhibition zones and the lack of further growth of both Gram-positive S. aureus and Gram-negative E. coli on the samples demonstrated the controlled release of everolimus, indicating the potential of the PDA layer in holding the everolimus release to the surrounding. Therefore, the study concludes that the formation of everolimus coating on biomaterial surfaces aided by the PDA layer, have significant potential in retarding bacterial colonies on medical devices.
... Within minutes, long-and short-range physicochemical interactions, such as Brownian motion, hydrophobic interactions, electrostatic interactions, and Van der Waals forces, followed by stronger molecular and cellular interactions result in the attraction, adsorption, and attachment of bacteria to PMMA surfaces. 16,17 These early bacterial colonizers proliferate and form multilayered cell clusters, embedded within their protective exopolysaccharide matrix. 15 Streptococcus species are the most common early colonizers and their presence and metabolites shift microenvironmental conditions and create attachment sites, which allow for the attachment and survival of later colonizers. ...
Article
Purpose: An interdisciplinary clinical review on denture stomatitis formulated by experts from prosthodontics, oral medicine, and oral microbiology is presented. The review outlines the etiopathogenesis, clinical features and management strategies of denture stomatitis from an interdisciplinary perspective. Materials and methods: English-language articles relating to the definition, incidence, gender distribution, geographical distribution, etiology, pathogenesis, symptoms, signs, treatment and prognosis of denture stomatitis were retrieved via electronic literature search. Relevant articles were summarized for this manuscript with a view towards providing pragmatic information. Results: Denture stomatitis represents a very common, multi-factorial infectious, inflammatory and hyperplastic condition which is primarily caused by poor oral hygiene, poor denture hygiene and full-time; mainly night-time denture wear, bringing about the emergence of advanced Candida-containing polymicrobial biofilms in close proximity to the host's mucosal tissues. Denture stomatitis shares clinically relevant associations with dental caries, periodontitis, median rhomboid glossitis, angular cheilitis, aspirational pneumonia and associated mortality. Conclusions: Effective, long-term treatment of denture stomatitis relies upon sustained patient-driven behavioral change which should focus on daily prosthesis-level cleaning and disinfection, removal of dentures at night, every night, engagement with professional denture maintenance, and when required, denture replacement. Anti-fungal medications offer limited benefit outside of short-term use due to the emergence of anti-fungal resistance. For frail, elderly, medically-compromised and nursing-home populations, treatment of denture stomatitis lowers the risk of aspirational pneumonia and associated mortality. This article is protected by copyright. All rights reserved.
... Bakteriyel yapışma; Van der Waals çekim kuvvetleri, Brown hareketi, yer çekimi kuvvetleri, elektrostatik yükler ve hidrofobik etkileşimler aracılığıyla hücrelerin yüzeye doğru ilk çekimiyle gerçekleşmektedir. [8] Biyofilm üretiminin bir sonraki fazı, serbest yüzen hücrelerin bir mikrokoloni düzenlemesi şeklinde yapışılacak yüzeyin üzerinde çoğalması ile karakterizedir. Bu aşamada, hücresel hareketlilik azalmakta ve ekzopolisakkarit üretimi, planktonik hücreleri ve besinleri çekmek için aktive edilmektedir. ...
... An important challenge for GBR materials applied in periodontitis is infection for that periodontitis is infectious diseases. Furthermore, application of GBR materials also could cause implant associated infections, which would exacerbate pre-existing infections and lead to progression of periodontitis [125]. GBR materials designed to be applied in periodontitis must be evaluated in more simulative conditions rather than been tested in frequently applied subcutaneous implanted model and cranial defect model. ...
Article
Full-text available
The anatomy of the oral and maxillofacial sites is complex, and bone defects caused by trauma, tumors, and inflammation in these zones are extremely difficult to repair. Among the most effective and reliable methods to attain osteogenesis, the guided bone regeneration (GBR) technique is extensively applied in defective oral and maxillofacial GBR. Furthermore, endowing biofunctions is crucial for GBR materials applied in repairing defective alveolar and maxillofacial bones. In this review, recent advances in designing and fabricating GBR materials applied in oral and maxillofacial sites are classified and discussed according to their biofunctions, including maintaining space for bone growth; facilitating the adhesion, migration, and proliferation of osteoblasts; facilitating the migration and differentiation of progenitor cells; promoting vascularization; providing immunoregulation to induce osteogenesis; suppressing infection; and effectively mimicking natural tissues using graded biomimetic materials. In addition, new processing strategies (e.g., 3D printing) and new design concepts (e.g., developing bone mimetic extracellular matrix niches and preparing scaffolds to suppress connective tissue to actively acquire space for bone regeneration), are particularly worthy of further study. In the future, GBR materials with richer biological functions are expected to be developed based on an in-depth understanding of the mechanism of bone-GBR-material interactions.
Chapter
Periprosthetic joint infection (PJI) is a devastating complication after total joint arthroplasty. The optimal PJI treatment depends on several factors such as presentation and chronicity of the infection, characteristics of the responsible microorganism, host health status, and overall surgeon/institutional experience Two-stage exchange arthroplasty is the preferred method for treating chronic PJI in North America. The first stage involves thorough debridement and removal of the infected prosthesis, followed by placement of an antibiotic-loaded cement spacer and treatment with targeted systemic antibiotics. Surgeons generally choose between placing either a static or an articulating spacer. Several options for spacer construction exist including spacer molds, preformed constructs, or hand-crafted devices. The second stage involves spacer removal and reimplantation with revision components. The choice of spacer type and design is dependent on several patient and surgeon-related factors. Infection eradication and improved function are the ultimate goals of two-stage exchange arthroplasty. Reported outcomes of this management option are acceptable. However, many important risk factors for treatment failure do exist. This chapter outlines the management of PJI with two-stage exchange arthroplasty with an emphasis on the diagnosis of PJI, surgical indications, surgical technique, and treatment outcomes.
Article
Full-text available
Blood-contacting medical devices such as biodegradable metallic bone implant materials are expected to show excellent hemocompatibility both in vitro and in vivo. Different approaches are being studied and used to modify biomaterial surfaces for enhanced biocompatibility and hemocompatibility. However, the composition of degradable biomaterial must address several drawbacks at once. Iron-reinforced zinc material was used as a metallic substrate with improved mechanical properties when compared with those of pure zinc. Poly(lactic) acid (PLA) or polyethylenimine (PEI) was selected as a polymeric matrix for further doping with antibiotic ciprofloxacin (CPR) and marine-sourced polysaccharide fucoidan (FU), which are known for their antibacterial and potential anticoagulant properties, respectively. Radiofrequency air plasma was employed to induce metallic/polymer-coated surface activation before further modification with FU/CPR. Sample surface morphology and composition were studied and evaluated (contact angle measurements, AFM, SEM, and FT-IR) along with the hemolysis ratio and platelet adhesion test. Successful doping of the polymer layer by FU/CRP was confirmed. While PEI induced severe hemolysis over 12%, the PLA-coated samples exhibited even lower hemolysis (∼2%) than uncoated samples while the uncoated samples showed the lowest platelet adhesion. Moreover, gradual antibiotic release from PLA determined by the electrochemical methods using screen-printed carbon electrodes was observed after 24, 48, and 72 h, making the PLA-coated zinc-based material an attractive candidate for biodegradable material design.
Article
Full-text available
Tricalcium phosphate (TCP) has gained attention due to its interconnected porous structures which promote fibrovascular invasion and bony replacement. Moreover, when gelatin is added and crosslinked with genipin (GGT), TCP exhibits robust biocompatibility and stability, making it an excellent bone substitute. In this study, we incorporated emodin and lumbrokinase (LK) into GGT to develop an antibacterial biomaterial. Emodin, derived from various plants, possesses antibacterial and anti-inflammatory properties. LK comprises proteolytic enzymes extracted from the earthworm Lumbricus rubellus and exhibits fibrinolytic activity, enabling it to dissolve biofilms. Additionally, LK stimulates osteoblast activity while inhibiting osteoclast differentiation. GGT was combined with emodin and lumbrokinase to produce the GGTELK composite. The biomedical effects of GGTELK were assessed through in vitro assays and an ex vivo bone defect model. The GGTELK composite demonstrated antibacterial properties, inhibiting the growth of S. aureus and reducing biofilm formation. Moreover, it exhibited anti-inflammatory effects by reducing the secretion of IL-6 in both in vivo cell experiments and the ex vivo model. Therefore, the GGTELK composite, with its stability, efficient degradation, biocompatibility, and anti-inflammatory function, is expected to serve as an ideal bone substitute.
Article
This review discussed the relationship among copper, human, and bacteria. Copper plays an important role in human immunity. Copper can boost human immune defense reactions at recommended intake level. The content mainly focused on copper antibacterial activity and copper antibacterial mechanisms. Conclusions stated that copper antibacterial activity is affected by copper homeostasis mechanisms in bacteria, adhesion, humidity, strain specificity, and manufacturing methods of antibacterial agents. For the preparation of particle antibacterial agents and surface antibacterial agents, this review discussed several manufacturing methods, such as sol-gel, cold spray, and biosynthesis belonging to chemical synthesis, physical synthesis, and biological synthesis, respectively. Sol-gel method contributes to the preparation of particle agents and surface agents. Cold spray technique is utilized in synthesis of surface copper agent. Biosynthesis is a novel technology which can be applied in nanoparticle agent preparation.
Article
The non‐covalent forces involved in bioadhesion involving flat surfaces, small particles, cells and bio‐polymers are: Lifshitz‐van der Waals (LW), polar (electron‐acceptor‐electron‐donor) or Lewis acid‐base (AB) and electrostatic (EL) forces. LW and AB forces are determined by: (a) direct contact angle measurement with at least three liquids on flat surfaces, or (b) wicking, by capillary rise measurements of at least three liquids along glass plates coated with a thin layer of small particles. This approach yields the LW and AB components of the surface tension, as well as the electron‐acceptor and the electron donor parameters of its AB‐component. Together, these yield the total apolar + polar interfacial energies involved in bioadhesion. EL interaction energies must be obtained separately, by electrokinetic methods. In bioadhesion, the importance of these forces usually is: AB > > LW > EL. The electron‐donor properties of cells and biopolymers are dominant; their electron‐acceptor parameters are negligible or zero. The degree of hydrophilicity of most materials can be expressed fairly precisely, as the interfacial free energy of attraction between the material and water; it consists principally of LW interactions. The interfacial free energy of interaction between biomaterials and synthetic surfaces, immersed in water is, for the greatest part, due to AB forces.The surface properties of biopolymers and cells usually are such that a maximum of adhesion should occur onto synthetic polymer surfaces with a surface tension of approximately 30 mJ rn. The adhesion of cells to clay particles is also studied: few cells will adhere to moderately hydrophilic'clay particles (hectorite), but all cells will tend to adhere to hydrophobic clay particles (talc). A minimum of cell adhesion occurs where a maximum of protein adsorption has taken place.
Article
In this study, a new and simple method is described to prepare wettability gradients on polymers by means of glow discharge in a partly shielded argon plasma. The surface characteristics of thus prepared gradients on low density polyethylene were determined by contact angle measurements and electron spectroscopy for chemical analysis (ESCA) and demonstrate that a chemical gradient of oxidized groups extends over a length of up to 4.5 cm, depending on the height of the plasma cover and the duration of the treatment. The chemical functionalities in which the oxygen was involved, however, were the same over the length of all gradients. Advancing water contact angles decrease from around 100° on the hydrophobic end to 40° on the hydrophilic end, with a contact angle hysteresis of around 40°. The steepest gradient was obtained with the cover at smaller distance above the polyethylene. Adsorption of albumin, fibrinogen and immunoglobulin G along the length of such a gradient surfaces increased with the distance from the hydrophobic end.
Article
Deep infection, the most serious local complication of total hip replacement, prompted a study of the records of 135 patients (137 hips) thus afflicted in a nationwide survey of Canada. Particular attention has been paid to the natural history of the infection, and the problems of diagnosis are described. Twenty-one patients died after the insertion, or removal, of the prosthesis, and of the survivors of the original 135 patients only eighteen have been able to retain the prosthesis without further problems with the wound. The remaining patients had the prosthesis removed, and most dry wounds. Certain suggestions are made on management. The advice that a second total hip prosthesis should be inserted after a deep infection of the first implant is not supported.
Article
Strains of indigenous Escherichia coli, Bacteroides, and Lactobacillus were isolated from the gastrointestinal tracts of specific pathogen-free (SPF) mice. Nonvaccinated SPF mice exhibited in their spleens low numbers of plaque-forming cells (PFC) and rosette-forming cells reacting with antigens of these andigenous bacteria. PFC reacting with these bacterial antigens were not detected in infant SPF mice until 7 days after birth. Compared with nonvaccinated controls, SPF mice vaccinated parenterally with indigenous E. coli or Bacteroides produced a moderate increase in the numbers of specific PFC. Thus, the SPF mouse is capable of responding immunologically after vaccination with microbes indigenous to its intestinal tract. However, more PFC reacting with homologous vaccine antigens were detected after parenteral vaccination of SPF mice with nonindigenous E. coli O127:B8, E.coli O14, or B. fragilis than after parenteral vaccination with indigenous E. coli or Bacteroides. Gnotobiotic mice orally monoassociated with these nonindigenous bacteria exhibited greater immune responses to antigens of the bacteria used to monoassociation than did gnotobiotes monoassociated with the indigenous microbes. The results are consistent with the hypothesis that mice are more responsive immunologically to antigens of nonindigenous bacteria than they are to antigens of certain microbes indigenous to their gastrointestinal tracts.
Article
Biocompatibility is concerned with the interactions that occur between biomaterials and host tissues. As foreign objects in that host tissue these materials may initiate several types of response. It has often been postulated that the immune response, by which the host normally defends itself against invasion by foreign organisms, can be involved in the response to biomaterials. This review discusses the mechanisms by which this could occur and the evidence that suggests the immune response is indeed of significance in biocompatibility.
Article
The adhesion and growth of two catheter-associated (O2K2 and O83K?) and two non catheter-associated (O111K58 and O157K-) uropathogenic Escherichia coli strains on glass, poly(methyl methacrylate) (PMMA), a negatively charged copolymer of MMA and methacrylic acid (MAA) and a positively charged copolymer of MMA and trimethylaminoethyl methacrylate chloride (TMAEMA-Cl) were studied. The solid surfaces were placed in a parallel plate perfusion system. After preadhesion of the bacteria onto the surfaces, growth was initiated by perfusing the system with MacConkey broth. Growth was measured by counting adherent bacteria as a function of time. Bacterial strains were characterized by means of water contact angle, microbial adhesion to hydrocarbon (MATH), anion exchange resin retention (ARR) and zeta potential measurements. Solid surfaces were characterized by means of water contact angle and zeta potential measurements. The catheter-associated strains had significantly higher water contact angles, zeta potentials and ARR values than the non catheter-associated strains. Non catheter-associated strains did not grow at the surfaces used. Catheter-associated strains did not grow at the positively charged surface but exhibited growth at the other surfaces. Strains grew more rapidly at surfaces with a relatively high negative zeta potential and a low water contact angle than at surfaces with a relatively low negative zeta potential and a high water contact angle. The growth of strain O2K2 on glass was significantly reduced when urine instead of MacConkey broth was used as perfusion medium.
Article
Sterile and endotoxin-free biomaterials commonly used in prosthetic devices (Dacron velour, woven Dacron, and Biomer polyurethane) and cotton (control material) were implanted intraperitoneally in mice with normal enteric flora. Intraperitoneal Biomer and woven Dacron became contaminated with 100 to 10,000 enteric bacteria, including Escherichia coli, Pseudomonas aeruginosa, enterococci, and staphylococci species, within 3 days; intraperitoneal cotton and Dacron velour were contaminated within 24 hours. Mesenteric lymph nodes showed parallel incidences of translocation. The peritoneal cavity became contaminated only if the biomaterial itself became contaminated. No bacterial overgrowth, perforation, or histologic changes in the bowel were found. Subcutaneous biomaterials remained sterile. Ingested fluorescent beads appeared in enterocytes, in lamina propria within macrophages, and in intraperitoneal biomaterials. The data suggest that intraperitoneal sterile reactive stimuli can induce bacterial translocation to the dense prosthesis directly through the intact normal bowel wall. One of the mechanisms seems to involve phagocytosis of particles and bacteria within the bowel wall that are then chemotactically attracted to nearby sites of inflammation.
Article
Clinically, Staphylococcus aureus appears to be the dominant organism associated with infected metal implants, whereas coagulase-negative staphylococcal strains are more frequently isolated from infected polymer implants. We reproduced this trend experimentally in vitro and in vivo. Discs of a titanium alloy, poly(methyl methacrylate) and ultra-high molecular weight polyethylene were exposed to a clinical isolate of Staphylococcus aureus or either of two strains of Staphylococcus epidermidis. Within 1 h Staphylococcus aureus was always the most rapid colonizer regardless of biomaterial. However, after 8 to 24 h, Staphylococcus aureus was present in higher numbers on metal and Staphylococcus epidermidis on polymers. Moreover, the exopolysaccharide produced by Staphylococcus epidermidis appeared to offer an effective protection against host defences in vivo.
Article
In this article, we will review some of the experimental evidence that supports the role of leukocytes in tissues injury. Our review will be divided into three major sections. As a background to subsequent sections, in the first section we will briefly review the various substances that can be released from leukocytes that could potentially contribute to tissue injury. In the second section, we will review studies that have implicated leukocytes as mediators of tissue injury, and in the final section we will review studies that have attempted to determine which specific leukocyte-derived substances might be responsible for leukocyte-induced tissue injury, and we will describe some of the specific cellular targets of these substances.