Skip directly to site content Skip directly to page options Skip directly to A-Z link Skip directly to A-Z link Skip directly to A-Z link
Issue Cover for Volume 12, Number 11—November 2006

Volume 12, Number 11—November 2006

[PDF - 7.81 MB - 187 pages]

Perspective

Prophylaxis and Treatment of Pregnant Women for Emerging Infections and Bioterrorism Emergencies [PDF - 182 KB - 7 pages]
J. Cono et al.

Emerging infectious disease outbreaks and bioterrorism attacks warrant urgent public health and medical responses. Response plans for these events may include use of medications and vaccines for which the effects on pregnant women and fetuses are unknown. Healthcare providers must be able to discuss the benefits and risks of these interventions with their pregnant patients. Recent experiences with outbreaks of severe acute respiratory syndrome, monkeypox, and anthrax, as well as response planning for bioterrorism and pandemic influenza, illustrate the challenges of making recommendations about treatment and prophylaxis for pregnant women. Understanding the physiology of pregnancy, the factors that influence the teratogenic potential of medications and vaccines, and the infection control measures that may stop an outbreak will aid planners in making recommendations for care of pregnant women during large-scale infectious disease emergencies.

EID Cono J, Cragan JD, Jamieson DJ, Rasmussen SA. Prophylaxis and Treatment of Pregnant Women for Emerging Infections and Bioterrorism Emergencies. Emerg Infect Dis. 2006;12(11):1631-1637. https://doi.org/10.3201/eid1211.060618
AMA Cono J, Cragan JD, Jamieson DJ, et al. Prophylaxis and Treatment of Pregnant Women for Emerging Infections and Bioterrorism Emergencies. Emerging Infectious Diseases. 2006;12(11):1631-1637. doi:10.3201/eid1211.060618.
APA Cono, J., Cragan, J. D., Jamieson, D. J., & Rasmussen, S. A. (2006). Prophylaxis and Treatment of Pregnant Women for Emerging Infections and Bioterrorism Emergencies. Emerging Infectious Diseases, 12(11), 1631-1637. https://doi.org/10.3201/eid1211.060618.

Emerging Infections and Pregnancy [PDF - 145 KB - 6 pages]
D. J. Jamieson et al.

A key component of the response to emerging infections is consideration of special populations, including pregnant women. Successful pregnancy depends on adaptation of the woman's immune system to tolerate a genetically foreign fetus. Although the immune system changes are not well understood, a shift from cell-mediated immunity toward humoral immunity is believed to occur. These immunologic changes may alter susceptibility to and severity of infectious diseases in pregnant women. For example, pregnancy may increase susceptibility to toxoplasmosis and listeriosis and may increase severity of illness and increase mortality rates from influenza and varicella. Compared with information about more conventional disease threats, information about emerging infectious diseases is quite limited. Pregnant women's altered response to infectious diseases should be considered when planning a response to emerging infectious disease threats.

EID Jamieson DJ, Theiler RN, Rasmussen SA. Emerging Infections and Pregnancy. Emerg Infect Dis. 2006;12(11):1638-1643. https://doi.org/10.3201/eid1211.060152
AMA Jamieson DJ, Theiler RN, Rasmussen SA. Emerging Infections and Pregnancy. Emerging Infectious Diseases. 2006;12(11):1638-1643. doi:10.3201/eid1211.060152.
APA Jamieson, D. J., Theiler, R. N., & Rasmussen, S. A. (2006). Emerging Infections and Pregnancy. Emerging Infectious Diseases, 12(11), 1638-1643. https://doi.org/10.3201/eid1211.060152.

Health Consequences of Child Marriage in Africa [PDF - 114 KB - 6 pages]
N. M. Nour

Despite international agreements and national laws, marriage of girls <18 years of age is common worldwide and affects millions. Child marriage is a human rights violation that prevents girls from obtaining an education, enjoying optimal health, bonding with others their own age, maturing, and ultimately choosing their own life partners. Child marriage is driven by poverty and has many effects on girls' health: increased risk for sexually transmitted diseases, cervical cancer, malaria, death during childbirth, and obstetric fistulas. Girls' offspring are at increased risk for premature birth and death as neonates, infants, or children. To stop child marriage, policies and programs must educate communities, raise awareness, engage local and religious leaders, involve parents, and empower girls through education and employment.

EID Nour NM. Health Consequences of Child Marriage in Africa. Emerg Infect Dis. 2006;12(11):1644-1649. https://doi.org/10.3201/eid1211.060510
AMA Nour NM. Health Consequences of Child Marriage in Africa. Emerging Infectious Diseases. 2006;12(11):1644-1649. doi:10.3201/eid1211.060510.
APA Nour, N. M. (2006). Health Consequences of Child Marriage in Africa. Emerging Infectious Diseases, 12(11), 1644-1649. https://doi.org/10.3201/eid1211.060510.

Anatidae Migration in the Western Palearctic and Spread of Highly Pathogenic Avian Influenza H5N1 Virus [PDF - 317 KB - 7 pages]
H. Yu et al.

During the second half of 2005, highly pathogenic avian influenza (HPAI) H5N1 virus spread rapidly from central Asia to eastern Europe. The relative roles of wild migratory birds and the poultry trade are still unclear, given that little is yet known about the range of virus hosts, precise movements of migratory birds, or routes of illegal poultry trade. We document and discuss the spread of the HPAI H5N1 virus in relation to species-specific flyways of Anatidae species (ducks, geese, and swans) and climate. We conclude that the spread of HPAI H5N1 virus from Russia and Kazakhstan to the Black Sea basin is consistent in space and time with the hypothesis that birds in the Anatidae family have seeded the virus along their autumn migration routes.

EID Yu H, Xiao X, Domenech J, Lubroth J, Martin V, Slingenbergh J. Anatidae Migration in the Western Palearctic and Spread of Highly Pathogenic Avian Influenza H5N1 Virus. Emerg Infect Dis. 2006;12(11):1650-1656. https://doi.org/10.3201/eid1211.060223
AMA Yu H, Xiao X, Domenech J, et al. Anatidae Migration in the Western Palearctic and Spread of Highly Pathogenic Avian Influenza H5N1 Virus. Emerging Infectious Diseases. 2006;12(11):1650-1656. doi:10.3201/eid1211.060223.
APA Yu, H., Xiao, X., Domenech, J., Lubroth, J., Martin, V., & Slingenbergh, J. (2006). Anatidae Migration in the Western Palearctic and Spread of Highly Pathogenic Avian Influenza H5N1 Virus. Emerging Infectious Diseases, 12(11), 1650-1656. https://doi.org/10.3201/eid1211.060223.

Review of Aerosol Transmission of Influenza A Virus [PDF - 179 KB - 6 pages]
R. Tellier

In theory, influenza viruses can be transmitted through aerosols, large droplets, or direct contact with secretions (or fomites). These 3 modes are not mutually exclusive. Published findings that support the occurrence of aerosol transmission were reviewed to assess the importance of this mode of transmission. Published evidence indicates that aerosol transmission of influenza can be an important mode of transmission, which has obvious implications for pandemic influenza planning and in particular for recommendations about the use of N95 respirators as part of personal protective equipment.

EID Tellier R. Review of Aerosol Transmission of Influenza A Virus. Emerg Infect Dis. 2006;12(11):1657-1662. https://doi.org/10.3201/eid1211.060426
AMA Tellier R. Review of Aerosol Transmission of Influenza A Virus. Emerging Infectious Diseases. 2006;12(11):1657-1662. doi:10.3201/eid1211.060426.
APA Tellier, R. (2006). Review of Aerosol Transmission of Influenza A Virus. Emerging Infectious Diseases, 12(11), 1657-1662. https://doi.org/10.3201/eid1211.060426.
Research

Susceptibility of North American Ducks and Gulls to H5N1 Highly Pathogenic Avian Influenza Viruses [PDF - 293 KB - 8 pages]
J. D. Brown et al.

Since 2002, H5N1 highly pathogenic avian influenza (HPAI) viruses have been associated with deaths in numerous wild avian species throughout Eurasia. We assessed the clinical response and extent and duration of viral shedding in 5 species of North American ducks and laughing gulls (Larus atricilla) after intranasal challenge with 2 Asian H5N1 HPAI viruses. Birds were challenged at ≈10 to 16 weeks of age, consistent with temporal peaks in virus prevalence and fall migration. All species were infected, but only wood ducks (Aix sponsa) and laughing gulls exhibited illness or died. Viral titers were higher in oropharyngeal swabs than in cloacal swabs. Duration of viral shedding (1–10 days) increased with severity of clinical disease. Both the hemagglutination-inhibition (HI) and agar gel precipitin (AGP) tests were able to detect postinoculation antibodies in surviving wood ducks and laughing gulls; the HI test was more sensitive than the AGP in the remaining 4 species

EID Brown JD, Stallknecht DE, Beck JR, Suarez DL, Swayne DE. Susceptibility of North American Ducks and Gulls to H5N1 Highly Pathogenic Avian Influenza Viruses. Emerg Infect Dis. 2006;12(11):1663-1670. https://doi.org/10.3201/eid1211.060652
AMA Brown JD, Stallknecht DE, Beck JR, et al. Susceptibility of North American Ducks and Gulls to H5N1 Highly Pathogenic Avian Influenza Viruses. Emerging Infectious Diseases. 2006;12(11):1663-1670. doi:10.3201/eid1211.060652.
APA Brown, J. D., Stallknecht, D. E., Beck, J. R., Suarez, D. L., & Swayne, D. E. (2006). Susceptibility of North American Ducks and Gulls to H5N1 Highly Pathogenic Avian Influenza Viruses. Emerging Infectious Diseases, 12(11), 1663-1670. https://doi.org/10.3201/eid1211.060652.

Targeted Social Distancing Designs for Pandemic Influenza [PDF - 661 KB - 11 pages]
R. J. Glass et al.

Targeted social distancing to mitigate pandemic influenza can be designed through simulation of influenza's spread within local community social contact networks. We demonstrate this design for a stylized community representative of a small town in the United States. The critical importance of children and teenagers in transmission of influenza is first identified and targeted. For influenza as infectious as 1957–58 Asian flu (≈50% infected), closing schools and keeping children and teenagers at home reduced the attack rate by >90%. For more infectious strains, or transmission that is less focused on the young, adults and the work environment must also be targeted. Tailored to specific communities across the world, such design would yield local defenses against a highly virulent strain in the absence of vaccine and antiviral drugs.

EID Glass RJ, Glass LM, Beyeler WE, Min HJ. Targeted Social Distancing Designs for Pandemic Influenza. Emerg Infect Dis. 2006;12(11):1671-1681. https://doi.org/10.3201/eid1211.060255
AMA Glass RJ, Glass LM, Beyeler WE, et al. Targeted Social Distancing Designs for Pandemic Influenza. Emerging Infectious Diseases. 2006;12(11):1671-1681. doi:10.3201/eid1211.060255.
APA Glass, R. J., Glass, L. M., Beyeler, W. E., & Min, H. J. (2006). Targeted Social Distancing Designs for Pandemic Influenza. Emerging Infectious Diseases, 12(11), 1671-1681. https://doi.org/10.3201/eid1211.060255.

Seroprevalence of Hepatitis E Virus Infection, Rural Southern People’s Republic of China [PDF - 190 KB - 7 pages]
R. Li et al.

Genotype 4 hepatitis E virus (HEV) is the dominant cause of hepatitis E in the People's Republic of China; swine are the principal reservoir. Our study was conducted in 8 rural communities of southern China, where families keep pigs near their homes. Phylogenetic analysis showed that 23 of 24 concurrent virus isolates from this region are genotype 4 strains. Among the study populations, immunoglobulin G anti-HEV seroprevalence accumulated with age at ≈1% per year for persons >60 years of age. After age 30 years, seroprevalence increased at higher rates for male than for female study participants. The overall seroprevalence was 43% (range 25%–66%) among the communities. Infection rates were higher for participants between 25 and 29 years of age. The results suggest that HEV infection probably has been endemic in southern China for <60 years, with swine being the principal reservoir of human HEV infection in recent years.

EID Li R, Ge S, Li Y, Zheng Y, Nong Y, Guo Q, et al. Seroprevalence of Hepatitis E Virus Infection, Rural Southern People’s Republic of China. Emerg Infect Dis. 2006;12(11):1682-1688. https://doi.org/10.3201/eid1211.060332
AMA Li R, Ge S, Li Y, et al. Seroprevalence of Hepatitis E Virus Infection, Rural Southern People’s Republic of China. Emerging Infectious Diseases. 2006;12(11):1682-1688. doi:10.3201/eid1211.060332.
APA Li, R., Ge, S., Li, Y., Zheng, Y., Nong, Y., Guo, Q....Xia, N. (2006). Seroprevalence of Hepatitis E Virus Infection, Rural Southern People’s Republic of China. Emerging Infectious Diseases, 12(11), 1682-1688. https://doi.org/10.3201/eid1211.060332.

Spatiotemporal Analysis of Invasive Meningococcal Disease, Germany [PDF - 177 KB - 7 pages]
J. Elias et al.

Meningococci can cause clusters of disease. Specimens from 1,616 patients in Germany obtained over 42 months were typed by serogrouping and sequence typing of PorA and FetA and yielded a highly diverse dataset (Simpson's index 0.963). A retrospective spatiotemporal scan statistic (SaTScan) was applied in an automated fashion to identify clusters for each finetype defined by serogroup variable region (VR) VR1 and VR2 of the PorA and VR of the FetA. A total of 26 significant clusters (p<0.05) were detected. On average, a cluster consisted of 2.6 patients. The median population in the geographic area of a cluster was 475,011, the median cluster duration was 4.0 days, and the proportion of cases in spatiotemporal clusters was 4.2%. The study exemplifies how the combination of molecular finetyping and spatiotemporal analysis can be used to assess an infectious disease in a large European country.

EID Elias J, Harmsen D, Claus H, Hellenbrand W, Frosch M, Vogel U. Spatiotemporal Analysis of Invasive Meningococcal Disease, Germany. Emerg Infect Dis. 2006;12(11):1689-1695. https://doi.org/10.3201/eid1211.060682
AMA Elias J, Harmsen D, Claus H, et al. Spatiotemporal Analysis of Invasive Meningococcal Disease, Germany. Emerging Infectious Diseases. 2006;12(11):1689-1695. doi:10.3201/eid1211.060682.
APA Elias, J., Harmsen, D., Claus, H., Hellenbrand, W., Frosch, M., & Vogel, U. (2006). Spatiotemporal Analysis of Invasive Meningococcal Disease, Germany. Emerging Infectious Diseases, 12(11), 1689-1695. https://doi.org/10.3201/eid1211.060682.

Schistosomiasis among Travelers: New Aspects of an Old Disease [PDF - 102 KB - 5 pages]
E. Schwartz et al.

Schistosomiasis is increasingly encountered among travelers returning from the tropics; signs and symptoms of travelers may differ from those of local populations. During 1993–2005, schistosomiasis was diagnosed in 137 Israeli travelers, most of whom were infected while in sub-Saharan Africa. Clinical findings compatible with acute schistosomiasis were recorded for 75 (66.4%) patients and included fever (71.3%), respiratory symptoms (42.9%), and cutaneous symptoms (45.2%). At time of physical examination, 42 patients (37.1%) still had symptoms of acute schistosomiasis, chronic schistosomiasis had developed in 23 (20.4%), and 48 (42.5%) were asymptomatic. Of patients who were initially asymptomatic, chronic schistosomiasis developed in 26%. Diagnosis was confirmed by serologic testing for 87.6% of patients, but schistosome ova were found in only 25.6%. We conclude that acute schistosomiasis is a major clinical problem among travelers, diagnostic and therapeutic options for acute schistosomiasis are limited, and asymptomatic travelers returning from schistosomiasis-endemic areas should be screened and treated.

EID Schwartz E, Artom G, Marva E, Assous MV, Rahav G, Schwartz E. Schistosomiasis among Travelers: New Aspects of an Old Disease. Emerg Infect Dis. 2006;12(11):1696-1700. https://doi.org/10.3201/eid1211.060340
AMA Schwartz E, Artom G, Marva E, et al. Schistosomiasis among Travelers: New Aspects of an Old Disease. Emerging Infectious Diseases. 2006;12(11):1696-1700. doi:10.3201/eid1211.060340.
APA Schwartz, E., Artom, G., Marva, E., Assous, M. V., Rahav, G., & Schwartz, E. (2006). Schistosomiasis among Travelers: New Aspects of an Old Disease. Emerging Infectious Diseases, 12(11), 1696-1700. https://doi.org/10.3201/eid1211.060340.

Gastroenteritis and Transmission of Helicobacter pylori Infection in Households [PDF - 130 KB - 8 pages]
S. Perry et al.

The mode of transmission of Helicobacter pylori infection is poorly characterized. In northern California, 2,752 household members were tested for H. pylori infection in serum or stool at a baseline visit and 3 months later. Among 1,752 person considered uninfected at baseline, 30 new infections (7 definite, 7 probable, and 16 possible) occurred, for an annual incidence of 7% overall and 21% in children <2 years of age. Exposure to an infected household member with gastroenteritis was associated with a 4.8-fold (95% confidence interval [CI] 1.4–17.1) increased risk for definite or probable new infection, with vomiting a greater risk factor (adjusted odds ratio [AOR] 6.3, CI 1.6–24.5) than diarrhea only (AOR 3.0, p = 0.65). Of probable or definite new infections, 75% were attributable to exposure to an infected person with gastroenteritis. Exposure to an H. pylori–infected person with gastroenteritis, particularly vomiting, markedly increased risk for new infection.

EID Perry S, Sanchez Md, Yang S, Haggerty TD, Hurst P, Perez-Perez G, et al. Gastroenteritis and Transmission of Helicobacter pylori Infection in Households. Emerg Infect Dis. 2006;12(11):1701-1708. https://doi.org/10.3201/eid1211.060086
AMA Perry S, Sanchez Md, Yang S, et al. Gastroenteritis and Transmission of Helicobacter pylori Infection in Households. Emerging Infectious Diseases. 2006;12(11):1701-1708. doi:10.3201/eid1211.060086.
APA Perry, S., Sanchez, M. d., Yang, S., Haggerty, T. D., Hurst, P., Perez-Perez, G....Parsonnet, J. (2006). Gastroenteritis and Transmission of Helicobacter pylori Infection in Households. Emerging Infectious Diseases, 12(11), 1701-1708. https://doi.org/10.3201/eid1211.060086.

Serotype Competence and Penicillin Resistance in Streptococcus pneumoniae [PDF - 138 KB - 6 pages]
Y. Hsieh et al.

From 2003 to 2005, we prospectively collected 118 isolates of pneumococci belonging to 7 serotypes to investigate their competence under the influence of the synthetic competence-stimulating peptides. The degree of competence of the various serotypes differed significantly. Serotype 6B had the highest competence, followed by serotypes 14, 19F, 9V, 23F, 3, and 18C. Isolates belonging to serotype 6B had greater genetic diversity than isolates belonging to serotype 3, which has high genetic clustering. Isolates belonging to serotypes 3 and 18C that were 100% sensitive to penicillin were significantly less competent than isolates belonging to serotypes 6B, 14, 19F, 9V, and 23F, which were frequently resistant to penicillin. Under the 7-valent pneumococcal conjugate vaccine program, enhanced molecular surveillance of virulent clones with higher competence to detect serotype switching will become more important.

EID Hsieh Y, Wang J, Lee W, Hsueh P, Shao P, Chang L, et al. Serotype Competence and Penicillin Resistance in Streptococcus pneumoniae. Emerg Infect Dis. 2006;12(11):1709-1714. https://doi.org/10.3201/eid1211.060414
AMA Hsieh Y, Wang J, Lee W, et al. Serotype Competence and Penicillin Resistance in Streptococcus pneumoniae. Emerging Infectious Diseases. 2006;12(11):1709-1714. doi:10.3201/eid1211.060414.
APA Hsieh, Y., Wang, J., Lee, W., Hsueh, P., Shao, P., Chang, L....Huang, L. (2006). Serotype Competence and Penicillin Resistance in Streptococcus pneumoniae. Emerging Infectious Diseases, 12(11), 1709-1714. https://doi.org/10.3201/eid1211.060414.

Staphylococcus aureus–associated Skin and Soft Tissue Infections in Ambulatory Care [PDF - 296 KB - 9 pages]
L. F. McCaig et al.

To describe the number and treatment of skin and soft tissue infections likely caused by Staphylococcus aureus in the United States, we analyzed data from the 1992–1994 and 2001–2003 National Ambulatory Medical Care Surveys and National Hospital Ambulatory Medical Care Surveys. Each year, data were reported by an average of 1,400 physicians, 230 outpatient departments, and 390 emergency departments for 30,000, 33,000, and 34,000 visits, respectively. During 2001–2003, the number of annual ambulatory care visits for skin and soft tissue infections was 11.6 million; the visit rate was 410.7 per 10,000 persons. During the study period, rates of overall and physician office visits did not differ; however, rates of visits to outpatient and emergency departments increased by 59% and 31%, respectively. This increase may reflect the emergence of community-acquired methicillin-resistant S. aureus infections.

EID McCaig LF, McDonald LC, Mandal S, Jernigan DB. Staphylococcus aureus–associated Skin and Soft Tissue Infections in Ambulatory Care. Emerg Infect Dis. 2006;12(11):1715-1723. https://doi.org/10.3201/eid1211.060190
AMA McCaig LF, McDonald LC, Mandal S, et al. Staphylococcus aureus–associated Skin and Soft Tissue Infections in Ambulatory Care. Emerging Infectious Diseases. 2006;12(11):1715-1723. doi:10.3201/eid1211.060190.
APA McCaig, L. F., McDonald, L. C., Mandal, S., & Jernigan, D. B. (2006). Staphylococcus aureus–associated Skin and Soft Tissue Infections in Ambulatory Care. Emerging Infectious Diseases, 12(11), 1715-1723. https://doi.org/10.3201/eid1211.060190.

Humans as Reservoir for Enterotoxin Gene–carrying Clostridium perfringens Type A [PDF - 243 KB - 6 pages]
A. Heikinheimo et al.

We found a prevalence of 18% for enterotoxin gene–carrying (cpe+) Clostridium perfringens in the feces of healthy food handlers by PCR and isolated the organism from 11 of 23 PCR-positive persons by using hydrophobic grid membrane filter-colony hybridization. Several different cpe genotypes were recovered. The prevalence was 3.7% for plasmidial IS1151-cpe, 2.9% for plasmidial IS1470-like-cpe, 0.7% for chromosomal IS1470-cpe, and 1.5% for unknown cpe genotype. Lateral spread of cpe between C. perfringens strains was evident because strains from the same person carried IS1470-like cpe but shared no genetic relatedness according to pulsed-field gel electrophoresis analysis. Our findings suggest that healthy humans serve as a rich reservoir for cpe+ C. perfringens type A and may play a role in the etiology of gastrointestinal diseases caused by this organism. The results also indicate that humans should be considered a risk factor for spread of C. perfringens type A food poisoning and that they are a possible source of contamination for C. perfringens type A food poisoning.

EID Heikinheimo A, Lindström M, Granum PE, Korkeala H. Humans as Reservoir for Enterotoxin Gene–carrying Clostridium perfringens Type A. Emerg Infect Dis. 2006;12(11):1724-1729. https://doi.org/10.3201/eid1211.060478
AMA Heikinheimo A, Lindström M, Granum PE, et al. Humans as Reservoir for Enterotoxin Gene–carrying Clostridium perfringens Type A. Emerging Infectious Diseases. 2006;12(11):1724-1729. doi:10.3201/eid1211.060478.
APA Heikinheimo, A., Lindström, M., Granum, P. E., & Korkeala, H. (2006). Humans as Reservoir for Enterotoxin Gene–carrying Clostridium perfringens Type A. Emerging Infectious Diseases, 12(11), 1724-1729. https://doi.org/10.3201/eid1211.060478.

Clostridium difficile PCR Ribotypes in Calves, Canada [PDF - 129 KB - 7 pages]
A. Rodriguez-Palacios et al.

We investigated Clostridium difficile in calves and the similarity between bovine and human C. difficile PCR ribotypes by conducting a case-control study of calves from 102 dairy farms in Canada. Fecal samples from 144 calves with diarrhea and 134 control calves were cultured for C. difficile and tested with an ELISA for C. difficile toxins A and B. C. difficile was isolated from 31 of 278 calves: 11 (7.6%) of 144 with diarrhea and 20 (14.9%) of 134 controls (p = 0.009). Toxins were detected in calf feces from 58 (56.8%) of 102 farms, 57 (39.6%) of 144 calves with diarrhea, and 28 (20.9%) of 134 controls (p = 0.0002). PCR ribotyping of 31 isolates showed 8 distinct patterns; 7 have been identified in humans, 2 of which have been associated with outbreaks of severe disease (PCR types 017 and 027). C. difficile may be associated with calf diarrhea, and cattle may be reservoirs of C. difficile for humans.

EID Rodriguez-Palacios A, Stämpfli HR, Duffield T, Peregrine AS, Trotz-Williams LA, Arroyo LG, et al. Clostridium difficile PCR Ribotypes in Calves, Canada. Emerg Infect Dis. 2006;12(11):1730-1736. https://doi.org/10.3201/eid1211.051581
AMA Rodriguez-Palacios A, Stämpfli HR, Duffield T, et al. Clostridium difficile PCR Ribotypes in Calves, Canada. Emerging Infectious Diseases. 2006;12(11):1730-1736. doi:10.3201/eid1211.051581.
APA Rodriguez-Palacios, A., Stämpfli, H. R., Duffield, T., Peregrine, A. S., Trotz-Williams, L. A., Arroyo, L. G....Weese, J. (2006). Clostridium difficile PCR Ribotypes in Calves, Canada. Emerging Infectious Diseases, 12(11), 1730-1736. https://doi.org/10.3201/eid1211.051581.
Dispatches

VEB-1 in Achromobacter xylosoxidans from Cystic Fibrosis Patient, France [PDF - 95 KB - 3 pages]
C. Neuwirth et al.

Multidrug-resistant Achromobacter xylosoxidans was recovered from the sputum of a patient with cystic fibrosis. The VEB-1 extended-spectrum β-lactamase was detected on a class 1 integron. This first report of a VEB-1–producing isolate in this population requires further investigation to determine its distribution.

EID Neuwirth C, Freby C, Ogier-Desserrey A, Perez-Martin S, Houzel A, Péchinot A, et al. VEB-1 in Achromobacter xylosoxidans from Cystic Fibrosis Patient, France. Emerg Infect Dis. 2006;12(11):1737-1739. https://doi.org/10.3201/eid1211.060143
AMA Neuwirth C, Freby C, Ogier-Desserrey A, et al. VEB-1 in Achromobacter xylosoxidans from Cystic Fibrosis Patient, France. Emerging Infectious Diseases. 2006;12(11):1737-1739. doi:10.3201/eid1211.060143.
APA Neuwirth, C., Freby, C., Ogier-Desserrey, A., Perez-Martin, S., Houzel, A., Péchinot, A....Siebor, E. (2006). VEB-1 in Achromobacter xylosoxidans from Cystic Fibrosis Patient, France. Emerging Infectious Diseases, 12(11), 1737-1739. https://doi.org/10.3201/eid1211.060143.

Molecular Characterization of Tickborne Relapsing Fever Borrelia, Israel [PDF - 183 KB - 4 pages]
M. V. Assous et al.

Blood samples from 18 tickborne relapsing fever (TBRF) patients and Ornithodoros tholozani specimens were tested with a Borrelia flaB-PCR. Results were positive for all patients and 2%–40% of ticks. A 7–amino acid gap characterized all 9 sequenced flagellin gene amplicons. By phylogenetic analysis, Israel TBRF Borrelia sequences clustered separately from American and African groups.

EID Assous MV, Wilamowski A, Bercovier H, Marva E. Molecular Characterization of Tickborne Relapsing Fever Borrelia, Israel. Emerg Infect Dis. 2006;12(11):1740-1743. https://doi.org/10.3201/eid1211.060715
AMA Assous MV, Wilamowski A, Bercovier H, et al. Molecular Characterization of Tickborne Relapsing Fever Borrelia, Israel. Emerging Infectious Diseases. 2006;12(11):1740-1743. doi:10.3201/eid1211.060715.
APA Assous, M. V., Wilamowski, A., Bercovier, H., & Marva, E. (2006). Molecular Characterization of Tickborne Relapsing Fever Borrelia, Israel. Emerging Infectious Diseases, 12(11), 1740-1743. https://doi.org/10.3201/eid1211.060715.

Fatal Avian Influenza A H5N1 in a Dog [PDF - 281 KB - 4 pages]
T. Songserm et al.

Avian influenza H5N1 virus is known to cross the species barrier and infect humans and felines. We report a fatal H5N1 infection in a dog following ingestion of an H5N1-infected duck during an outbreak in Thailand in 2004. With new reports of H5N1 virus continuing across Asia, Europe, and Africa, this finding highlights the need for monitoring of domestic animals during outbreaks.

EID Songserm T, Amonsin A, Jam-on R, Sae-Heng N, Pariyothorn N, Payungporn S, et al. Fatal Avian Influenza A H5N1 in a Dog. Emerg Infect Dis. 2006;12(11):1744-1747. https://doi.org/10.3201/eid1211.060542
AMA Songserm T, Amonsin A, Jam-on R, et al. Fatal Avian Influenza A H5N1 in a Dog. Emerging Infectious Diseases. 2006;12(11):1744-1747. doi:10.3201/eid1211.060542.
APA Songserm, T., Amonsin, A., Jam-on, R., Sae-Heng, N., Pariyothorn, N., Payungporn, S....Poovorawan, Y. (2006). Fatal Avian Influenza A H5N1 in a Dog. Emerging Infectious Diseases, 12(11), 1744-1747. https://doi.org/10.3201/eid1211.060542.

Reemergence of Dengue Virus Type 4, French Antilles and French Guiana, 2004–2005 [PDF - 198 KB - 4 pages]
P. Dussart et al.

After 10 years of absence, dengue virus type 4 (DENV-4) has recently reemerged in Martinique, Guadeloupe, and French Guiana. Phylogenetic analyses of strains isolated from 2004 to 2005 showed that they belong to DENV-4 genotype II, but to a different cluster than strains isolated from 1993 to 1995.

EID Dussart P, Lavergne A, Lagathu G, Lacoste V, Martial J, Morvan J, et al. Reemergence of Dengue Virus Type 4, French Antilles and French Guiana, 2004–2005. Emerg Infect Dis. 2006;12(11):1748-1751. https://doi.org/10.3201/eid1211.060339
AMA Dussart P, Lavergne A, Lagathu G, et al. Reemergence of Dengue Virus Type 4, French Antilles and French Guiana, 2004–2005. Emerging Infectious Diseases. 2006;12(11):1748-1751. doi:10.3201/eid1211.060339.
APA Dussart, P., Lavergne, A., Lagathu, G., Lacoste, V., Martial, J., Morvan, J....Cesaire, R. (2006). Reemergence of Dengue Virus Type 4, French Antilles and French Guiana, 2004–2005. Emerging Infectious Diseases, 12(11), 1748-1751. https://doi.org/10.3201/eid1211.060339.

Genotype III Saint Louis Encephalitis Virus Outbreak, Argentina, 2005 [PDF - 96 KB - 3 pages]
L. A. Diaz et al.

Twenty-six years after it was last detected, Saint Louis encephalitis virus (SLEV) genotype III reemerged in 2005 in Córdoba, Argentina, where it caused an outbreak. Two genotype III SLEV strains were isolated from Culex quinquefasciatus. A 71.43% prevalence for neutralizing antibodies was found in domestic fowl in the homestead of a patient with encephalitis.

EID Diaz LA, Ré V, Almirón WR, Farías A, Vázquez A, Sanchez-Seco MP, et al. Genotype III Saint Louis Encephalitis Virus Outbreak, Argentina, 2005. Emerg Infect Dis. 2006;12(11):1752-1754. https://doi.org/10.3201/eid1211.060486
AMA Diaz LA, Ré V, Almirón WR, et al. Genotype III Saint Louis Encephalitis Virus Outbreak, Argentina, 2005. Emerging Infectious Diseases. 2006;12(11):1752-1754. doi:10.3201/eid1211.060486.
APA Diaz, L. A., Ré, V., Almirón, W. R., Farías, A., Vázquez, A., Sanchez-Seco, M. P....Contigiani, M. (2006). Genotype III Saint Louis Encephalitis Virus Outbreak, Argentina, 2005. Emerging Infectious Diseases, 12(11), 1752-1754. https://doi.org/10.3201/eid1211.060486.

Human Parainfluenza Type 4 Infections, Canada [PDF - 216 KB - 4 pages]
M. Vachon et al.

During the fall/winter season of 2004–05, we found 9 respiratory specimens positive for human parainfluenza virus type 4 (HPIV-4) in our laboratory (43% of all HPIVs) from patients with mild to moderate respiratory illnesses. Sequencing studies identified 8 different HPIV-4A strains and 1 HPIV-4B strain.

EID Vachon M, Dionne N, Leblanc É, Moisan D, Bergeron MG, Boivin G. Human Parainfluenza Type 4 Infections, Canada. Emerg Infect Dis. 2006;12(11):1755-1758. https://doi.org/10.3201/eid1211.060196
AMA Vachon M, Dionne N, Leblanc É, et al. Human Parainfluenza Type 4 Infections, Canada. Emerging Infectious Diseases. 2006;12(11):1755-1758. doi:10.3201/eid1211.060196.
APA Vachon, M., Dionne, N., Leblanc, É., Moisan, D., Bergeron, M. G., & Boivin, G. (2006). Human Parainfluenza Type 4 Infections, Canada. Emerging Infectious Diseases, 12(11), 1755-1758. https://doi.org/10.3201/eid1211.060196.

Methicillin-Resistant Staphylococcus aureus at Canoe Camp [PDF - 77 KB - 3 pages]
K. Como-Sabetti et al.

We investigated a cluster of community-associated methicillin-resistant Staphylococcus aureus infections among persons at a wilderness canoe camp. Isolates from the investigation had identical profiles for susceptibility, pulsed-field gel electrophoresis, and toxins. Participants in activities that involve skin injury, person-to-person contact, and inadequate hygiene are at increased risk for methicillin-resistant S. aureus infections.

EID Como-Sabetti K, Harriman K, Juni B, Westbrook A, Cebelinski E, Boxrud D, et al. Methicillin-Resistant Staphylococcus aureus at Canoe Camp. Emerg Infect Dis. 2006;12(11):1759-1761. https://doi.org/10.3201/eid1211.060363
AMA Como-Sabetti K, Harriman K, Juni B, et al. Methicillin-Resistant Staphylococcus aureus at Canoe Camp. Emerging Infectious Diseases. 2006;12(11):1759-1761. doi:10.3201/eid1211.060363.
APA Como-Sabetti, K., Harriman, K., Juni, B., Westbrook, A., Cebelinski, E., Boxrud, D....Lynfield, R. (2006). Methicillin-Resistant Staphylococcus aureus at Canoe Camp. Emerging Infectious Diseases, 12(11), 1759-1761. https://doi.org/10.3201/eid1211.060363.

Knowledge, Attitudes, and Practices of Avian Influenza, Poultry Workers, Italy [PDF - 219 KB - 4 pages]
R. Abbate et al.

We asked Italian poultry workers about knowledge, attitudes, and practices regarding avian influenza. It was perceived to be a low occupational hazard, and wearing protective equipment and handwashing were not routine practices. Knowledge of transmission and preventive measures should be improved. Employers and health professionals should provide more effective information.

EID Abbate R, Di Giuseppe G, Marinelli P, Angelillo IF. Knowledge, Attitudes, and Practices of Avian Influenza, Poultry Workers, Italy. Emerg Infect Dis. 2006;12(11):1762-1765. https://doi.org/10.3201/eid1211.060671
AMA Abbate R, Di Giuseppe G, Marinelli P, et al. Knowledge, Attitudes, and Practices of Avian Influenza, Poultry Workers, Italy. Emerging Infectious Diseases. 2006;12(11):1762-1765. doi:10.3201/eid1211.060671.
APA Abbate, R., Di Giuseppe, G., Marinelli, P., & Angelillo, I. F. (2006). Knowledge, Attitudes, and Practices of Avian Influenza, Poultry Workers, Italy. Emerging Infectious Diseases, 12(11), 1762-1765. https://doi.org/10.3201/eid1211.060671.

Avian Influenza H5N1 Screening of Intensive Care Unit Patients with Community-acquired Pneumonia [PDF - 205 KB - 4 pages]
A. Apisarnthanarak et al.

From February 1, 2005, to January 31, 2006, we screened 115 adults for avian influenza (H5N1) and influenza A if admitted to an intensive care unit with pneumonia. Using reverse transcription-PCR, viral culture, and serologic testing for anti-H5 antibody, we identified 8 (7%) patients with influenza A (H3N2); none had H5N1. Estimated costs for H5N1 screening were $7,375.

EID Apisarnthanarak A, Puthavathana P, Kitphati R, Thavatsupha P, Chittaganpitch M, Auewarakul P, et al. Avian Influenza H5N1 Screening of Intensive Care Unit Patients with Community-acquired Pneumonia. Emerg Infect Dis. 2006;12(11):1766-1769. https://doi.org/10.3201/eid1211.060443
AMA Apisarnthanarak A, Puthavathana P, Kitphati R, et al. Avian Influenza H5N1 Screening of Intensive Care Unit Patients with Community-acquired Pneumonia. Emerging Infectious Diseases. 2006;12(11):1766-1769. doi:10.3201/eid1211.060443.
APA Apisarnthanarak, A., Puthavathana, P., Kitphati, R., Thavatsupha, P., Chittaganpitch, M., Auewarakul, P....Mundy, L. M. (2006). Avian Influenza H5N1 Screening of Intensive Care Unit Patients with Community-acquired Pneumonia. Emerging Infectious Diseases, 12(11), 1766-1769. https://doi.org/10.3201/eid1211.060443.

Transplacental Chikungunya Virus Antibody Kinetics, Thailand [PDF - 96 KB - 3 pages]
V. Watanaveeradej et al.

Antibodies to chikungunya virus were detected by hemagglutination-inhibition assay in 33.6% of 2,000 infants' cord sera at delivery. Follow-up of 24 seropositive infants showed that the half-life of antibody persistence was 35.5 days. Chikungunya virus infection is common in Thailand, and routine use of diagnostic assays is needed.

EID Watanaveeradej V, Endy TP, Simasathien S, Kerdpanich A, Polprasert N, Aree C, et al. Transplacental Chikungunya Virus Antibody Kinetics, Thailand. Emerg Infect Dis. 2006;12(11):1770-1772. https://doi.org/10.3201/eid1211.051560
AMA Watanaveeradej V, Endy TP, Simasathien S, et al. Transplacental Chikungunya Virus Antibody Kinetics, Thailand. Emerging Infectious Diseases. 2006;12(11):1770-1772. doi:10.3201/eid1211.051560.
APA Watanaveeradej, V., Endy, T. P., Simasathien, S., Kerdpanich, A., Polprasert, N., Aree, C....Nisalak, A. (2006). Transplacental Chikungunya Virus Antibody Kinetics, Thailand. Emerging Infectious Diseases, 12(11), 1770-1772. https://doi.org/10.3201/eid1211.051560.

Food Markets with Live Birds as Source of Avian Influenza [PDF - 101 KB - 3 pages]
X. Tang et al.
EID Tang X, Di B, Zhou D, Zheng B, Jing H, Lin Y, et al. Food Markets with Live Birds as Source of Avian Influenza. Emerg Infect Dis. 2006;12(11):1773-1775. https://doi.org/10.3201/eid1211.060675
AMA Tang X, Di B, Zhou D, et al. Food Markets with Live Birds as Source of Avian Influenza. Emerging Infectious Diseases. 2006;12(11):1773-1775. doi:10.3201/eid1211.060675.
APA Tang, X., Di, B., Zhou, D., Zheng, B., Jing, H., Lin, Y....Xu, J. (2006). Food Markets with Live Birds as Source of Avian Influenza. Emerging Infectious Diseases, 12(11), 1773-1775. https://doi.org/10.3201/eid1211.060675.

Recurrent Tuberculosis and Exogenous Reinfection, Shanghai, China [PDF - 84 KB - 3 pages]
G. Shen et al.

Of 52 patients with recurrent tuberculosis in Shanghai, People's Republic of China, 32 (61.5%) had isolates in which genotype patterns of Mycobacterium tuberculosis differed between first and second episodes. This result indicates that exogenous reinfection is common in an area with a high incidence of tuberculosis.

EID Shen G, Xue Z, Shen X, Sun B, Gui X, Shen M, et al. Recurrent Tuberculosis and Exogenous Reinfection, Shanghai, China. Emerg Infect Dis. 2006;12(11):1776-1778. https://doi.org/10.3201/eid1211.051207
AMA Shen G, Xue Z, Shen X, et al. Recurrent Tuberculosis and Exogenous Reinfection, Shanghai, China. Emerging Infectious Diseases. 2006;12(11):1776-1778. doi:10.3201/eid1211.051207.
APA Shen, G., Xue, Z., Shen, X., Sun, B., Gui, X., Shen, M....Gao, Q. (2006). Recurrent Tuberculosis and Exogenous Reinfection, Shanghai, China. Emerging Infectious Diseases, 12(11), 1776-1778. https://doi.org/10.3201/eid1211.051207.

Identical Genotype B3 Sequences from Measles Patients in 4 Countries, 2005 [PDF - 130 KB - 3 pages]
J. Rota et al.

Surveillance of measles virus detected an epidemiologic link between a refugee from Kenya and a Dutch tourist in New Jersey, USA. Identical genotype B3 sequences from patients with contemporaneous cases in the United States, Canada, and Mexico in November and December 2005 indicate that Kenya was likely to have been the common source of virus.

EID Rota J, Lowe L, Rota PA, Bellini W, Redd S, Dayan G, et al. Identical Genotype B3 Sequences from Measles Patients in 4 Countries, 2005. Emerg Infect Dis. 2006;12(11):1779-1781. https://doi.org/10.3201/eid1211.060635
AMA Rota J, Lowe L, Rota PA, et al. Identical Genotype B3 Sequences from Measles Patients in 4 Countries, 2005. Emerging Infectious Diseases. 2006;12(11):1779-1781. doi:10.3201/eid1211.060635.
APA Rota, J., Lowe, L., Rota, P. A., Bellini, W., Redd, S., Dayan, G....Santibanez, S. (2006). Identical Genotype B3 Sequences from Measles Patients in 4 Countries, 2005. Emerging Infectious Diseases, 12(11), 1779-1781. https://doi.org/10.3201/eid1211.060635.
Letters

Panton-Valentine Leukocidin–producing Staphylococcus aureus [PDF - 47 KB - 2 pages]
A. Adler et al.
EID Adler A, Temper V, Block CS, Abramson N, Moses AE. Panton-Valentine Leukocidin–producing Staphylococcus aureus. Emerg Infect Dis. 2006;12(11):1789-1790. https://doi.org/10.3201/eid1211.060726
AMA Adler A, Temper V, Block CS, et al. Panton-Valentine Leukocidin–producing Staphylococcus aureus. Emerging Infectious Diseases. 2006;12(11):1789-1790. doi:10.3201/eid1211.060726.
APA Adler, A., Temper, V., Block, C. S., Abramson, N., & Moses, A. E. (2006). Panton-Valentine Leukocidin–producing Staphylococcus aureus. Emerging Infectious Diseases, 12(11), 1789-1790. https://doi.org/10.3201/eid1211.060726.

Chikungunya Fever, Hong Kong [PDF - 52 KB - 3 pages]
N. Lee et al.
EID Lee N, Wong CK, Lam WY, Wong A, Lim W, Lam CW, et al. Chikungunya Fever, Hong Kong. Emerg Infect Dis. 2006;12(11):1790-1792. https://doi.org/10.3201/eid1211.060574
AMA Lee N, Wong CK, Lam WY, et al. Chikungunya Fever, Hong Kong. Emerging Infectious Diseases. 2006;12(11):1790-1792. doi:10.3201/eid1211.060574.
APA Lee, N., Wong, C. K., Lam, W. Y., Wong, A., Lim, W., Lam, C. W....Tang, J. W. (2006). Chikungunya Fever, Hong Kong. Emerging Infectious Diseases, 12(11), 1790-1792. https://doi.org/10.3201/eid1211.060574.

Screening Laboratory Requests [PDF - 48 KB - 2 pages]
C. A. Petti et al.
EID Petti CA, Polage CR, Hillyard DR. Screening Laboratory Requests. Emerg Infect Dis. 2006;12(11):1792-1793. https://doi.org/10.3201/eid1211.060711
AMA Petti CA, Polage CR, Hillyard DR. Screening Laboratory Requests. Emerging Infectious Diseases. 2006;12(11):1792-1793. doi:10.3201/eid1211.060711.
APA Petti, C. A., Polage, C. R., & Hillyard, D. R. (2006). Screening Laboratory Requests. Emerging Infectious Diseases, 12(11), 1792-1793. https://doi.org/10.3201/eid1211.060711.

Malaria Outbreak in Troops Returning from French Guiana [PDF - 61 KB - 2 pages]
C. Verret et al.
EID Verret C, Cabianca B, Haus-Cheymol R, Lafille J, Loran-Haranqui G, Spiegel A. Malaria Outbreak in Troops Returning from French Guiana. Emerg Infect Dis. 2006;12(11):1794-1795. https://doi.org/10.3201/eid1211.060530
AMA Verret C, Cabianca B, Haus-Cheymol R, et al. Malaria Outbreak in Troops Returning from French Guiana. Emerging Infectious Diseases. 2006;12(11):1794-1795. doi:10.3201/eid1211.060530.
APA Verret, C., Cabianca, B., Haus-Cheymol, R., Lafille, J., Loran-Haranqui, G., & Spiegel, A. (2006). Malaria Outbreak in Troops Returning from French Guiana. Emerging Infectious Diseases, 12(11), 1794-1795. https://doi.org/10.3201/eid1211.060530.

Plasmodium vivax Malaria Relapses after Primaquine Prophylaxis [PDF - 39 KB - 2 pages]
P. Reddy and J. P. Flaherty
EID Reddy P, Flaherty JP. Plasmodium vivax Malaria Relapses after Primaquine Prophylaxis. Emerg Infect Dis. 2006;12(11):1795-1796. https://doi.org/10.3201/eid1211.060613
AMA Reddy P, Flaherty JP. Plasmodium vivax Malaria Relapses after Primaquine Prophylaxis. Emerging Infectious Diseases. 2006;12(11):1795-1796. doi:10.3201/eid1211.060613.
APA Reddy, P., & Flaherty, J. P. (2006). Plasmodium vivax Malaria Relapses after Primaquine Prophylaxis. Emerging Infectious Diseases, 12(11), 1795-1796. https://doi.org/10.3201/eid1211.060613.

Avian Influenza and US TV News [PDF - 35 KB - 2 pages]
B. G. Southwell et al.
EID Southwell BG, Hwang Y, Torres A. Avian Influenza and US TV News. Emerg Infect Dis. 2006;12(11):1797-1798. https://doi.org/10.3201/eid1211.060672
AMA Southwell BG, Hwang Y, Torres A. Avian Influenza and US TV News. Emerging Infectious Diseases. 2006;12(11):1797-1798. doi:10.3201/eid1211.060672.
APA Southwell, B. G., Hwang, Y., & Torres, A. (2006). Avian Influenza and US TV News. Emerging Infectious Diseases, 12(11), 1797-1798. https://doi.org/10.3201/eid1211.060672.

Resistance to Dihydroartemisinin [PDF - 34 KB - 2 pages]
S. Cojean et al.
EID Cojean S, Hubert V, Le Bras J, Durand R. Resistance to Dihydroartemisinin. Emerg Infect Dis. 2006;12(11):1798-1799. https://doi.org/10.3201/eid1211.060903
AMA Cojean S, Hubert V, Le Bras J, et al. Resistance to Dihydroartemisinin. Emerging Infectious Diseases. 2006;12(11):1798-1799. doi:10.3201/eid1211.060903.
APA Cojean, S., Hubert, V., Le Bras, J., & Durand, R. (2006). Resistance to Dihydroartemisinin. Emerging Infectious Diseases, 12(11), 1798-1799. https://doi.org/10.3201/eid1211.060903.

Real-time PCR for Francisella tularensis Types A and B [PDF - 62 KB - 3 pages]
K. J. Kugeler et al.
EID Kugeler KJ, Pappert R, Zhou Y, Petersen JM. Real-time PCR for Francisella tularensis Types A and B. Emerg Infect Dis. 2006;12(11):1799-1801. https://doi.org/10.3201/eid1211.060629
AMA Kugeler KJ, Pappert R, Zhou Y, et al. Real-time PCR for Francisella tularensis Types A and B. Emerging Infectious Diseases. 2006;12(11):1799-1801. doi:10.3201/eid1211.060629.
APA Kugeler, K. J., Pappert, R., Zhou, Y., & Petersen, J. M. (2006). Real-time PCR for Francisella tularensis Types A and B. Emerging Infectious Diseases, 12(11), 1799-1801. https://doi.org/10.3201/eid1211.060629.

Concurrent Plasmodium vivax Malaria and Dengue [PDF - 21 KB - 1 page]
S. Deresinski
EID Deresinski S. Concurrent Plasmodium vivax Malaria and Dengue. Emerg Infect Dis. 2006;12(11):1802. https://doi.org/10.3201/eid1211.060341
AMA Deresinski S. Concurrent Plasmodium vivax Malaria and Dengue. Emerging Infectious Diseases. 2006;12(11):1802. doi:10.3201/eid1211.060341.
APA Deresinski, S. (2006). Concurrent Plasmodium vivax Malaria and Dengue. Emerging Infectious Diseases, 12(11), 1802. https://doi.org/10.3201/eid1211.060341.

Viruses from Nonhuman Primates [PDF - 59 KB - 2 pages]
R. A. Vilchez
EID Vilchez RA. Viruses from Nonhuman Primates. Emerg Infect Dis. 2006;12(11):1802-1803. https://doi.org/10.3201/eid1211.060659
AMA Vilchez RA. Viruses from Nonhuman Primates. Emerging Infectious Diseases. 2006;12(11):1802-1803. doi:10.3201/eid1211.060659.
APA Vilchez, R. A. (2006). Viruses from Nonhuman Primates. Emerging Infectious Diseases, 12(11), 1802-1803. https://doi.org/10.3201/eid1211.060659.

Rickettsia parkeri in Uruguay [PDF - 39 KB - 2 pages]
R. C. Pacheco et al.
EID Pacheco RC, Venzal JM, Richtzenhain LJ, Labruna MB. Rickettsia parkeri in Uruguay. Emerg Infect Dis. 2006;12(11):1804-1805. https://doi.org/10.3201/eid1211.060577
AMA Pacheco RC, Venzal JM, Richtzenhain LJ, et al. Rickettsia parkeri in Uruguay. Emerging Infectious Diseases. 2006;12(11):1804-1805. doi:10.3201/eid1211.060577.
APA Pacheco, R. C., Venzal, J. M., Richtzenhain, L. J., & Labruna, M. B. (2006). Rickettsia parkeri in Uruguay. Emerging Infectious Diseases, 12(11), 1804-1805. https://doi.org/10.3201/eid1211.060577.

Influenza-related Death Rates for Pregnant Women
P. Mortimer
EID Mortimer P. Influenza-related Death Rates for Pregnant Women. Emerg Infect Dis. 2006;12(11):1805-1806. https://doi.org/10.3201/eid1211.061071
AMA Mortimer P. Influenza-related Death Rates for Pregnant Women. Emerging Infectious Diseases. 2006;12(11):1805-1806. doi:10.3201/eid1211.061071.
APA Mortimer, P. (2006). Influenza-related Death Rates for Pregnant Women. Emerging Infectious Diseases, 12(11), 1805-1806. https://doi.org/10.3201/eid1211.061071.
Another Dimension

Sexual Health in Art and Science [PDF - 287 KB - 7 pages]
S. Semaan et al.
EID Semaan S, Des Jarlais DC, Bice S. Sexual Health in Art and Science. Emerg Infect Dis. 2006;12(11):1782-1788. https://doi.org/10.3201/eid1211.ad1211
AMA Semaan S, Des Jarlais DC, Bice S. Sexual Health in Art and Science. Emerging Infectious Diseases. 2006;12(11):1782-1788. doi:10.3201/eid1211.ad1211.
APA Semaan, S., Des Jarlais, D. C., & Bice, S. (2006). Sexual Health in Art and Science. Emerging Infectious Diseases, 12(11), 1782-1788. https://doi.org/10.3201/eid1211.ad1211.
Books and Media

OIE/FAO International Scientific Conference on Avian Influenza [PDF - 15 KB - 1 page]
N. Marano
EID Marano N. OIE/FAO International Scientific Conference on Avian Influenza. Emerg Infect Dis. 2006;12(11):1807. https://doi.org/10.3201/eid1211.061013
AMA Marano N. OIE/FAO International Scientific Conference on Avian Influenza. Emerging Infectious Diseases. 2006;12(11):1807. doi:10.3201/eid1211.061013.
APA Marano, N. (2006). OIE/FAO International Scientific Conference on Avian Influenza. Emerging Infectious Diseases, 12(11), 1807. https://doi.org/10.3201/eid1211.061013.
About the Cover

Women Caring for Children in “the Floating World” [PDF - 59 KB - 2 pages]
P. Potter
EID Potter P. Women Caring for Children in “the Floating World”. Emerg Infect Dis. 2006;12(11):1808-1809. https://doi.org/10.3201/eid1211.ac1211
AMA Potter P. Women Caring for Children in “the Floating World”. Emerging Infectious Diseases. 2006;12(11):1808-1809. doi:10.3201/eid1211.ac1211.
APA Potter, P. (2006). Women Caring for Children in “the Floating World”. Emerging Infectious Diseases, 12(11), 1808-1809. https://doi.org/10.3201/eid1211.ac1211.
Etymologia

Etymologia: chikungunya [PDF - 49 KB - 1 page]
EID Etymologia: chikungunya. Emerg Infect Dis. 2006;12(11):1772. https://doi.org/10.3201/eid1211.et1211
AMA Etymologia: chikungunya. Emerging Infectious Diseases. 2006;12(11):1772. doi:10.3201/eid1211.et1211.
APA (2006). Etymologia: chikungunya. Emerging Infectious Diseases, 12(11), 1772. https://doi.org/10.3201/eid1211.et1211.
Conference Summaries

The Second International Conference on Women and Infectious Diseases
M. McDonald and S. Romero-Steiner

Methicillin-Resistant Staphylococcus aureus as Community Pathogen
J. A. Jernigan et al.
Corrections

Correction: Vol. 12, No. 10 [PDF - 14 KB - 1 page]
EID Correction: Vol. 12, No. 10. Emerg Infect Dis. 2006;12(11):1805. https://doi.org/10.3201/eid1211.c11211
AMA Correction: Vol. 12, No. 10. Emerging Infectious Diseases. 2006;12(11):1805. doi:10.3201/eid1211.c11211.
APA (2006). Correction: Vol. 12, No. 10. Emerging Infectious Diseases, 12(11), 1805. https://doi.org/10.3201/eid1211.c11211.
Page created: November 02, 2011
Page updated: November 02, 2011
Page reviewed: November 02, 2011
The conclusions, findings, and opinions expressed by authors contributing to this journal do not necessarily reflect the official position of the U.S. Department of Health and Human Services, the Public Health Service, the Centers for Disease Control and Prevention, or the authors' affiliated institutions. Use of trade names is for identification only and does not imply endorsement by any of the groups named above.
edit_01 ScholarOne Submission Portal
Issue Select
GO
GO

Spotlight Topics

 

Get Email Updates

To receive email updates about this page, enter your email address:

file_external