You are on page 1of 54

Infectious Diseases of the Horse

Diagnosis pathology management and


public health 1st Edition Jh Van Der
Kolk
Visit to download the full and correct content document:
https://textbookfull.com/product/infectious-diseases-of-the-horse-diagnosis-pathology-
management-and-public-health-1st-edition-jh-van-der-kolk/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

Advances in Microbiology, Infectious Diseases and


Public Health: Volume 14 Gianfranco Donelli

https://textbookfull.com/product/advances-in-microbiology-
infectious-diseases-and-public-health-volume-14-gianfranco-
donelli/

Advances in Microbiology Infectious Diseases and Public


Health Volume 1 1st Edition Gianfranco Donelli (Eds.)

https://textbookfull.com/product/advances-in-microbiology-
infectious-diseases-and-public-health-volume-1-1st-edition-
gianfranco-donelli-eds/

Advances in Microbiology Infectious Diseases and Public


Health Volume 2 1st Edition Gianfranco Donelli (Eds.)

https://textbookfull.com/product/advances-in-microbiology-
infectious-diseases-and-public-health-volume-2-1st-edition-
gianfranco-donelli-eds/

Advances in Microbiology, Infectious Diseases and


Public Health: Volume 7 1st Edition Gianfranco Donelli
(Eds.)

https://textbookfull.com/product/advances-in-microbiology-
infectious-diseases-and-public-health-volume-7-1st-edition-
gianfranco-donelli-eds/
Emergency Management of Infectious Diseases Rachel L.
Chin

https://textbookfull.com/product/emergency-management-of-
infectious-diseases-rachel-l-chin/

Fungal Biofilms and related infections Advances in


Microbiology Infectious Diseases and Public Health
Volume 3 1st Edition Christine Imbert (Eds.)

https://textbookfull.com/product/fungal-biofilms-and-related-
infections-advances-in-microbiology-infectious-diseases-and-
public-health-volume-3-1st-edition-christine-imbert-eds/

Emerging and Re-emerging Viral Infections: Advances in


Microbiology, Infectious Diseases and Public Health
Volume 6 1st Edition Giovanni Rezza

https://textbookfull.com/product/emerging-and-re-emerging-viral-
infections-advances-in-microbiology-infectious-diseases-and-
public-health-volume-6-1st-edition-giovanni-rezza/

Pythium Diagnosis Diseases and Management 1st Edition


Mahendra Rai (Editor)

https://textbookfull.com/product/pythium-diagnosis-diseases-and-
management-1st-edition-mahendra-rai-editor/

Infectious Diseases Emergencies 1st Edition Bissonette

https://textbookfull.com/product/infectious-diseases-
emergencies-1st-edition-bissonette/
INFECTIOUS
DISEASES of the
HORSE Diagnosis, pathology,
management, and
public health

J. H. van der Kolk


DVM, PhD, dipl ECEIM
Department of Equine Sciences–Medicine
Faculty of Veterinary Medicine
Utrecht University, the Netherlands

E. J. B. Veldhuis Kroeze
DVM, dipl ECVP
Department of Pathobiology
Faculty of Veterinary Medicine
Utrecht University, the Netherlands

MANSON PUBLISHING / THE VETERINARY PRESS


Copyright © 2013 Manson Publishing Ltd
ISBN: 978-1-84076-165-8

All rights reserved. No part of this publication may be reproduced, stored in a retrieval system or transmitted
in any form or by any means without the written permission of the copyright holder or in accordance with
the provisions of the Copyright Act 1956 (as amended), or under the terms of any licence permitting limited
copying issued by the Copyright Licensing Agency, 33–34 Alfred Place, London WC1E 7DP, UK.

Any person who does any unauthorized act in relation to this publication may be liable to criminal
prosecution and civil claims for damages.

A CIP catalogue record for this book is available from the British Library.

For full details of all Manson Publishing Ltd titles please write to:
Manson Publishing Ltd, 73 Corringham Road, London NW11 7DL, UK.
Tel: +44(0)20 8905 5150
Fax: +44(0)20 8201 9233
Email: manson@mansonpublishing.com
Website: www.mansonpublishing.com

Commissioning editor: Jill Northcott


Project manager: Kate Nardoni
Copy editor: Ruth Maxwell
Layout: DiacriTech, Chennai, India
Colour reproduction: Tenon & Polert Colour Scanning Ltd, Hong Kong
Printed by: Grafos SA, Barcelona, Spain
CONTENTS
Introduction 5 Actinomyces spp. 90
Abbreviations 6 Dermatophilus congolensis: ‘rain scald’ or
streptotrichosis 92
Chapter 1 Corynebacterium pseudotuberculosis:
‘pigeon fever’ 94
Bacterial diseases 9
Mycobacterium spp.: tuberculosis 97
Nocardia spp. 99
Anaplasma phagocytophilum:
equine anaplasmosis 9 Rhodococcus equi: ‘rattles’ 100
Neorickettsia risticii: Potomac horse fever 12 Chlamydophila spp. 106
Bartonella henselae: bartonellosis 15 Borrelia burgdorferi: Lyme disease 108
Brucella spp.: brucellosis 16 Leptospira interrogans: leptospirosis 112
Burkholderia mallei: ‘glanders’ 18 Bacteroidaceae and Fusobacteriaceae 118
Burkholderia pseudomallei: melioidosis 20 Botryomycosis 120
Bordetella bronchiseptica 22
Taylorella equigenitalis: contagious Chapter 2
equine metritis 24 Viral diseases 123
Francisella tularensis: tularemia 26
Legionella pneumophila 27 Equine adenovirus 123
Coxiella burnetii: Q fever 28 Equine herpesvirus 126
Moraxella spp. 28 Suid herpesvirus 1 133
Escherichia coli 30 Bovine and equine papillomavirus 134
Salmonella spp.: salmonellosis 34 Horsepox virus 138
Pasteurella spp. 37 West Nile virus/Kunjin virus 140
Yersinia enterocolitica: yersiniosis 39 Japanese encephalitis virus/Murray Valley
Actinobacillus lignieresii: actinobacillosis encephalitis virus 143
(‘wooden tongue’) 40 Equine arteritis virus 144
Actinobacillus equuli: ‘sleepy foal disease’ 42 Equine influenza virus 148
Lawsonia intracellularis: equine proliferative Hendra virus 152
enteropathy 44 Borna disease virus 154
Campylobacter spp. 48 Equine rhinitis virus 156
Helicobacter equorum 49 African horse sickness virus 158
Clostridium botulinum: botulism (‘shaker Equine infectious anaemia virus: ‘swamp fever’ 162
foal disease’) 50
Rotavirus 164
Clostridium difficile 55
Rabies 166
Clostridium perfringens 58
Vesicular stomatitis virus 168
Clostridium piliforme: Tyzzer’s disease 62
Eastern equine encephalomyelitis virus 170
Clostridium tetani: tetanus 64
Western equine encephalomyelitis virus 172
Mycoplasma spp. 66
Venezuelan equine encephalomyelitis virus 174
Erysipelothrix rhusiopathiae 68
Getah virus and Ross River virus:
Bacillus anthracis: anthrax 70 Ross River virus and Sagiyama virus 176
Listeria monocytogenes: listeriosis 73
Methicillin-resistant Staphylococcus aureus 75
Streptococcus equi subsp. equi: ‘strangles’ 78
Streptococcus equi subsp. zooepidemicus 85
4

Chapter 3 Chapter 6
Protozoal diseases 178 Helmintic diseases 217

Klossiella equi 178 Fasciola hepatica 217


Sarcocystis neurona/Neospora hughesi: Anoplocephala spp. 224
equine protozoal myeloencephalitis (EPM) 180 Echinococcus equinus 228
Cryptosporidium parvum: cryptosporidiosis 184 Strongyloides westeri 232
Eimeria leuckarti 186 Halicephalobus gingivalis 234
Babesia caballi/Theileria equi: Strongylus spp. 236
babesiosis/piroplasmosis 188
Cyathostomum spp. 242
Giardia duodenalis 192
Dictyocaulus arnfieldi 248
Trypanosoma brucei evansi/T. b. equiperdum:
trypanosomosis 194 Parelaphostrongylus tenuis 249
Parascaris equorum 250
Chapter 4 Oxyuris equi 254
Fungal diseases 197 Probstmayria vivipara 258
Thelazia lacrymalis 258
Invasive mycoses 197 Setaria equina 260
Histoplasma capsulatum var. farciminosum:
equine epizootic lymphangitis/ Appendices 262
histoplasmosis/pseudofarcy 206

Appendix 1
Chapter 5
Differential diagnoses 262
Ectoparasitical diseases 208
Appendix 2
(Potential) Zoonoses 264
Gasterophilus spp. 208
Appendix 3
Mites 212
Clinical pathology 265
Lice 215

References 279
Index 327
5

INTRODUCTION
In equine medicine one of the most important In order to support clinicians, a list of
areas is the field of infectious diseases. This field is differential diagnoses has been provided in
very dynamic and ever evolving with emerging Appendix 1. Furthermore, Appendix 2 has been
and fading diseases. Many professionals are provided in an attempt to update the current view
dedicated to equine infectious diseases ranging on zoonotic aspects of equine infectious diseases.
from clinicians via laboratory diagnosticians to Appendix 3 emphasizes the importance of clinical
pathologists. This book is the outcome of close pathology in the diagnosis of infectious diseases.
collaboration between a clinician and a The authors hope that this book will be helpful
pathologist and as such positively affected the for anyone dealing with equine infectious diseases
selection of colour plates provided. Rapid and suggestions to improve future issues are more
development of molecular biology techniques has than welcome.
greatly improved diagnostic possibilities in equine
infectious diseases, and facilitates epidemiological We sincerely acknowledge the contributions of
as well as zoonotic studies. In this book the M. Aleman, C.M. Butler, A. van Dijk, G.C.M.
majority of equine infectious diseases are arranged Grinwis, E. Gruys, M. Heinrichs, D. Kersten,
based on the various microbes and parasites B. Malmhagen, K. Matiasek, G. Uilenberg,
involved, using Fauquet et al. (2005) for the E. Smiet, E. Teske, and V.M. van der Veen.
classification of viruses, Garrity et al. (2004) for
the classification of the prokaryotes, and Kassai References and further reading
(1999) for the classification of the helminths. In Coles EH 1986. Veterinary Clinical Pathology,
the individual sections the opportunities available 4th edn. WB Saunders, London.
for diagnosis of various causative agents using Fauquet CM, Mayo MA, Maniloff J,
molecular biology have been described. However, Desselberger U, Ball LA (eds) 2005. Virus
these opportunities are usually limited by the Taxonomy. 8th Report of the International
options provided by local diagnostic laboratories Committee on Taxonomy of Viruses.
and of course they should be contacted prior to Academic Press, Elsevier, Amsterdam.
sample submission. Nevertheless, the mere Garrity GM, Bell JA, Lilburn TG 2004.
presence of a microbe and/or parasite in or on an Taxonomic outline of the prokaryotes. In:
animal cannot be considered adequate evidence Bergey’s Manual of Systematic Bacteriology,
that it is the aetiological agent of a disease that 2nd edn. Springer, New York.
may exist. Diagnostic aids must be used to Kassai T 1999. Veterinary Helminthology.
supplement, not supplant, clinical observations. Butterworth-Heinemann, Oxford.

DISCLAIMER
The advice and information given in this book
are believed to be true and accurate at the time of
going to press. However, not all drugs,
formulations, and devices are currently available
in all countries, and readers are advised to check
local availability and prescribing regimens.
6

ABBREVIATIONS
ABL Australian bat lyssavirus EMG electromyography
ABV avian bornavirus EPA epidemic polyarthritis
ADV Aujeszky’s disease virus EPE equine proliferative enteropathy
AGID agar gel immunodiffusion EPM equine protozoal myeloencephalitis
AHS African horse sickness ERAV equine rhinitis A virus
AHSV African horse sickness virus ERBV equine rhinitis B virus
AI antibody index ETBF enterotoxigenic Bacteroides fragilis
AIDS aquired immune deficiency syndrome ExPEC extraintestinal pathogenic
AST aspartate aminotransferase Escherichia coli
BAL bronchoalveolar lavage FAT fluorescent antibody test
BCG bacillus Calmette–Guérin FEI Fédération Equestre Internationale
BDV Borna disease virus FMDV foot-and-mouth disease virus
bid twice daily γ-GT γ-glutamyl transferase
BoNT botulinum neurotoxin GALT gut-associated lymphoid tissue
BPV bovine papillomavirus GGT gamma-glutamyltransferase
BW body weight GI gastrointestinal
CA-MRSA community-associated methicillin- GLDH glutamate dehydrogenase
resistant Staphylococcus aureus HA-MRSA hospital-associated methicillin-resistant
CDC complement-dependent cytotoxicity Staphylococcus aureus
(assay) H&E haematoxylin and eosin
CEM contagious equine metritis HeV Hendra virus
CF complement fixation HI haemagglutination inhibition
CFT complement fixation test HIV human immunodeficiency virus
CK creatine kinase HJV Highlands J virus
CNF cytotoxic necrotizing factor HYPP hyperkalaemic periodic paralysis
CNS central nervous system IAD inflammatory airway disease
CPXV cowpox virus IFA immunofluorescence assay
CSF cerebrospinal fluid IFAT indirect fluorescent antibody test
CT computed tomography IFT immunofluorescence test
CTA cell cytotoxicity assay IgG(T) immune globulin G induced by tetanus
DDSP dorsal displacement of the soft palate toxoid
EAV equine arteritis virus IM intramuscular
EcPV equine papillomavirus IPMA immunoperoxidase monolayer assay
EDTA ethylenediaminetetraacetic acid IV intravenous
EEV equine encephalomyelitis virus JEV Japanese encephalitis virus
EEEV eastern equine encephalomyelitis virus KUN Kunjin virus
EGS equine grass sickness LAMP loop-mediated isothermal amplification
EHV equine herpesvirus LDH lactate dehydrogenase
EIA equine infectious anaemia, enzyme LPS lipopolysaccharide
immunoassay LTR long terminal repeat
EIAV equine infectious anaemia virus MAC IgM antibody capture
EIPH exercise-induced pulmonary MAT microscopic agglutination test
haemorrhage MIC minimum inhibitory concentration
EL (equine) epizootic lymphangitis MLST multilocus sequence typing
(c)ELISA (competitive) enzyme-linked MPXV monkeypox virus
immunosorbent assay
MRI magnetic resonance imaging
EM electron microscopy
7

MRSA methicillin-resistant Staphylococcus SCCmec staphylococcal cassette chromosome


aureus element carrying the mecA gene
MVA modified vaccinia Ankara SCID severe combined immune deficiency
MVE Murray Valley encephalitis (virus) SDS-PAGE sodium dodecyl sulphate
NA North America polyacrylamide gel electrophoresis
NASBA nucleic acid sequence based SHI synergistic haemolysis inhibition
amplification sid once a day
NiV Nipah virus SNT serum neutralization test
NSAID nonsteroidal anti-inflammatory drug spa encoding gene of protein A
OPV orthopoxvirus SRAP sequence-related amplified
PAGE polyacrylamide gel electrophoresis polymorphism
PAS periodic acid-Schiff SRH single radial haemolysis
PCR polymerase chain reaction SSCP single-strand conformation
polymorphism
PDD proventricular dilatation disease
TB tuberculosis
PEP post-exposure prophylaxis
TCE transarterial coil embolization
PFGE pulsed-field gel electrophoresis
TMP/S trimethoprim-potentiated sulphonamide
PFU plaque-forming unit
TMP/SDZ trimethoprim/sulphadiazine
PMT Pasteurella multocida toxin
USUV Usutu virus
PO per os
VACV vaccinia virus
PPIA pituitary pars intermedia adenoma
VEE Venezuelan equine encephalomyelitis
PRNT plaque reduction neutralization test
VEEV Venezuelan equine encephalomyelitis
PRV pseudorabies virus virus
RAO recurrent airway obstruction VN virus neutralization
RAPD random amplified polymorphic DNA VSIV vesicular stomatitis Indiana virus
RFLP restriction fragment length VSNJV vesicular stomatitis New Jersey virus
polymorphism
VSV vesicular stomatitis virus
RLB reverse line blot
WBC white blood cell
RRV Ross River virus
WEEV western equine encephalomyelitis virus
RT reverse transcription
WNV West Nile virus
SC subcutaneous
This page intentionally left blank
9

Chapter 1
Bacterial diseases

Anaplasma phagocytophilum: EQUINE Epidemiology


ANAPLASMOSIS Equine anaplasmosis was first described in the USA
Phylum BXII Proteobacteria in 1969 (Gribble 1969), and has since been reported
Class I Alphaproteobacteria/Order II in other countries, including Switzerland, Sweden,
Rickettsiales/Family II Anaplasmataceae/Genus I France, Germany, Italy, the UK, the Czech Republic,
Anaplasma and the Netherlands (Gerhards et al. 1987, Butler et
al. 2008, Zeman & Jahn 2009). Most infections
Definition/Overview develop during the late fall, winter, and spring
Equine anaplasmosis is a noncontagious infectious (Madigan & Gribble 1987). I. ricinus is one of the
disease of horses caused by Anaplasma vectors of A. phagocytophilum in Europe, in which
phagocytophilum (formerly named Ehrlichia rates of infection range from 1.9 to 34%. In 1997
phagocytophila and Ehrlichia equi) identified as only 0.4% of equine blood samples examined were
emerging in Europe (Vorou et al. 2007). found positive for antibodies to A. phagocytophilum
in the Latium region (Lillini et al. 2006). However,
Aetiology the rate of A. phagocytophilum antibody prevalence
Equine anaplasmosis is caused by the obligate in healthy horses on USA farms enzootic for
intracellular bacterium A. phagocytophilum. Cross- equine anaplasmosis can be as high as 10%
species differences in pathogenicity and ecologically (Madigan et al. 1990), whereas 9.8% of horses with
separate strains within this bacterial species appear fever of unknown origin tested positive for A.
to exist (Franzén et al. 2005, Foley et al. 2009), as phagocytophilum in the Netherlands (Butler et al.
two unique genetic variants infecting horses in the 2008).
Czech Republic were identified (Zeman & Jahn Transmission and propagation of
2009). Horses inoculated with the human-derived A. phagocytophilum occur in large mammals such
A. phagocytophilum agent results in clinical disease as horses, cattle, sheep, goats, dogs, and cats. Small
largely indistinguishable from equine anaplasmosis mammals and not ticks are the reservoirs
(Madigan et al. 1995). The mode of transmission is of anaplasmosis (Lillini et al. 2006). Roe deer
unknown (but it is most likely a tick), although co- are the main reservoir for A. phagocytophilum
infection with Borrelia burgdorferi is attributed to in central Europe and Scandinavia, with a high
their common vector. Ticks of the Ixodes ricinus seroprevalence of about 95% and a variable rate of
complex also act as vectors in the spread of B. polymerase chain reaction (PCR)-proven infection
burgdorferi and co-infections of A. phagocytophilum ranging from 12.5% in the Czech Republic to
and B. burgdorferi have been confirmed in horses 85.6% in Slovenia (Skarphedinsson et al. 2005).
(Chang et al. 2000, Magnarelli et al. 2000). The role of migrating birds in long-range tick
transfer may be important since the same A.
phagocytophilum gene sequences were detected in
infected ticks on migrating birds and in humans and
domestic animals in Sweden (Bjoersdorff et al.
2001).
10 Bacterial diseases

1 2

1 Clinical signs
seen in equine
anaplasmosis 2 Equine anaplasmosis. The integument is irregular due to
include distal generalized urticaria or hives (variably sized oedematous
limb oedema. bumps) especially apparent on thorax, neck, and proximal
extremities. A hypersensitivity reaction is implicated; this
feverish horse proved positive for Anaplasma
3 phagocytophilum.

3 Granulocytic anaplasmosis (ehrlichiosis). Equine


blood smear. The central neutrophil contains a cytoplasmic
ring-shaped inclusion consistent with Anaplasma
phagocytophilum. Inclusions may be detected in granulocytes
and are polymorphic, round, irregular to ring-shaped,
ranging from 0.75 to 3.5 μm in diameter. Round to ovoid
morulae (2.5–3.5 μm in diameter) are composed of small
granules. Single initial bodies measure approximately 0.5 μm
in diameter. (May–Grünwald–Giemsa stain.)

Incubation period disease can be self-limiting when untreated, and the


The incubation period in experimentally infected clinical signs abated and disappeared without
horses varies from 1 to 9 days (Stannard et al. 1969, specific treatment 7–14 days after onset of the
Franzén et al. 2005). One of two horses receiving disease (Gerhards et al. 1987, Gribble 1969). By 22
high dosage of infective blood (20 × 106 infected days after infection, in one study, all abnormal signs
neutrophils) died suddenly and unexpectedly 2 days associated with equine anaplasmosis had fully
into clinical illness (Franzén et al. 2005). abated in all surviving horses (Franzén et al. 2005).
However, infection with A. phagocytophilum can
Clinical presentation persist in the horse for at least 129 days although the
Clinical signs include high fever, depression, continued presence of the organism is not associated
inappetence, petechiation, icterus, ataxia, with detectable clinical or pathological abnormalities
rhabdomyolysis, and distal limb oedema (1) (Franzén et al. 2009). It is unclear whether
associated with lymphopenia, neutropenia, horses younger than 3–4 years of age generally
thrombocytopenia, and anaemia (Gribble experience less severe clinical disease (Gribble 1969,
1969, Madigan & Gribble 1987, Gerhards et al. Madigan & Gribble 1987, Butler et al. 2008).
1987, Madigan 1993, Franzén et al. 2005, Butler et Occasionally euthanasia is required because of
al. 2008, Hilton et al. 2008). Extensive urticaria may deterioration despite treatment (Butler et al. 2008).
also be associated with equine anaplasmosis (2). The
Bacterial diseases 11

4 5

4, 5 Granulocytic anaplasmosis (ehrlichiosis), cytology specimen from a blood smear containing several small bacterial
polymorphic cytoplasmic morulae (4, arrowhead) or single initial bodies (5, arrows) within neutrophils. Anaplasma
phagocytophilum. (May–Grünwald–Giemsa stain. Bars 10 μm.)

6 7

6, 7 Granulocytic anaplasmosis (ehrlichiosis), cytology specimens from a blood smear with infected neutrophils, containing
an intracytoplasmic ring form (6, arrow) and clustered initial bodies (7, arrowhead) of Anaplasma phagocytophilum.
(May–Grünwald-Giemsa stain. Bars 20 μm.)

Differential diagnosis 8
Clinical signs are similar to those caused
by infections with other pathogens such as
B. burgdorferi, B. caballi, Theileria equi, equine
herpesvirus, equine infectious anaemia virus, equine
arteritis virus, viral encephalitides and
Leptospiraceae (Butler et al. 2008).

Diagnosis
Diagnosis of equine anaplasmosis is usually based on
the detection of characteristic cytoplasmic inclusion
bodies in peripheral blood (3–7); either morulae or
elementary bodies are seen. In the neutrophilic and
occasionally eosinophilic granulocytes on a Wright–
Giemsa- or haematoxylin and eosin (H&E)-stained 8 Granulocytic anaplasmosis (ehrlichiosis), cytology specimen
smear of peripheral blood (8) obtained during days from a blood smear with three infected neutrophils each
3–5 of fever during peak ehrlichiaemia (Gribble containing different forms of A. phagocytophilum inclusions.
1969, Madigan & Gribble 1987, Madigan (May–Grünwald–Giemsa stain. Bar 20 μm.)
12 Bacterial diseases

1993). Morulae (<4 μm in diameter) consist Neorickettsia risticii: POTOMAC HORSE


of elementary bodies (<1 μm in diameter). FEVER
Microscopic interpretation of a buffy coat smear of Phylum BXII Proteobacteria
H&E-stained peripheral blood is a sensitive Class I Alphaproteobacteria/Order II
and practical diagnostic tool for the Rickettsiales/Family II Anaplasmataceae/Genus
veterinarian considering possible infection with V Neorickettsia
A. phagocytophilum in horses with pyrexia, but
some cases may require PCR testing for diagnosis Definition/Overview
(Butler et al. 2008, Hilton et al. 2008). In addition, Potomac horse fever (also known as equine
the PCR signal was consistently detected 2–3 days monocytic ehrlichiosis) is an acute and potentially
before appearance of clinical signs and persisted 4– fatal equine disease associated with depression,
9 days beyond abatement of clinical signs, whereas anorexia, fever, dehydration, laminitis, abortion, and
diagnostic inclusion bodies (varying from 0.5–16% watery diarrhoea (Holland et al. 1985, Rikihisa et
of neutrophils) were first noted on average 2.6±1.5 al. 1985) caused by Neorickettsia risticii (formerly
days after onset of fever (Franzén et al. 2005). Ehrlichia risticii).
Horses seroconverted by 12–16 days
after inoculation, reaching maximal indirect Aetiology
immunofluorescence assay (IFA) titres (up to N. risticii is an obligate intracellular bacterium of the
1:5,120) within 3–7 days from when seropositivity Anaplasmataceae family in the order Rickettsiales.
was identified (Franzén et al. 2005). The indirect The organism has an unique affinity for monocytes
fluorescent antibody titre to A. phagocytophilum and during the course of the disease and among
persists for approximately 300 days after horses, monocyte counts are variable, but they
inoculation of the organism (Nyindo et al. 1978). increase to 13% in some horses (Dutta et al. 1988).
Characteristic cytoplasmic inclusion bodies (either
Pathology morulae or elementary bodies) occur in the
Macroscopically, oedema of the ventrum and limbs monocytes from peripheral blood during peak
may be present including subcutaneous petechiae. ehrlichiaemia. Morulae (less than 4 μm in diameter)
Histologically there is evidence of vasculitides in the consist of elementary bodies (less than 1 μm in
affected subcutis (Jubb et al. 2007). During fever diameter) (Holland et al. 1985). The complete
rickettsial inclusions can be detected in granulocytes genome sequence of N. risticii consists of a single
of a blood smear, and are polymorphic, round, circular chromosome of 879, 977 base pairs and
irregular to ring-shaped, ranging from 0.75 to 3.5 encodes 38 RNA species and 898 proteins.
μm in diameter. Round to ovoid morulae (2.5–3.5 Comparison with its closely related human pathogen
μm in diameter) are composed of small granules. N. sennetsu showed that 758 (88.2%) of protein-
Single initial bodies measure approximately 0.5 μm coding genes are conserved between N. risticii and
in diameter (Jubb et al. 2007). N. sennetsu (Lin et al. 2009).

Management/Treatment Epidemiology
The treatment of choice is oxytetracycline 7 mg/kg It has been shown that the trematode Acanthatrium
BW IV sid for 3–7 days to hasten recovery and oregonense is a natural reservoir and probably a
alleviate clinical signs (Madigan & Gribble 1987). vector of N. risticii, as N. risticii is vertically
Clinical immunity in experimental horses was shown transmitted (from adult to egg) in A. oregonense
to last 2 years (Gribble 1969). (Gibson et al. 2005). In addition, caddisflies were
reported as second intermediate hosts of N. risticii-
Public health significance infected trematodes by carrying infected
Although five Anaplasmataceae members, including metacercariae (Madigan et al. 2000, Mott et al.
A. phagocytophilum, E. chaffeensis, E. ewingii, E. 2002). Furthermore, N. risticii can also be
canis, and Neorickettsia sennetsu infect humans, transmitted horizontally from trematode to bats
only the first three species have been investigated (Gibson et al. 2005). N. risticii has not been reported
fully. All forms of human ehrlichiosis share many outside the USA.
clinical and laboratory manifestations, including
fever, headache, myalgia and malaise,
thrombocytopenia, leucopenia, and indices of
hepatic injury (Dumler et al. 2007).
Bacterial diseases 13

Incubation period 9
The incubation period varies from 3 to 9 days.
Major clinical and haematological features of
induced N. risticii infection are biphasic increase in
rectal temperature (with peak increases to 38.9°C
and 39.3°C on post-inoculation days 5 and 12,
respectively), depression, anorexia, decreased white
blood cell (WBC) count (maximal decrease of 47%
on post-inoculation day 12), and diarrhoea from
post-inoculation days 14 to 18. Increased WBC
count was an inconsistent feature with a maximal
increase of 52% on post-inoculation day 20 (Dutta
et al. 1988).

Clinical presentation
Under field conditions, N. risticii infection is 9 Diarrhoea develops in the majority of horses suffering from
characterized by increased rectal temperature, anorexia, Potomac horse fever.
depression, leucopenia, and then diarrhoea (9).
Occasionally horses develop profound ileus and
severe colic (10) (Whitlock & Palmer 1986, Palmer 10
et al. 1986). Diarrhoea developed in 73% of horses
and mortality was 9% (Dutta et al. 1988). Laminitis
and limb oedema are often seen as a sequel to N.
risticii infection, and mortality is 10–20%
(Whitlock & Palmer 1986). However, clinically
undetectable infections exist (Ristic et al. 1986). In
utero infection (Dawson et al. 1987) has also been
reported as well as abortion (81 days post-infection)
with recovery of the organism from the bone
marrow of a fetus on the 200th day of gestation
(Long et al. 1992).

Differential diagnosis
The differential diagnosis includes various causes of
acute diarrhoea (see p. 263).

10 Colic can be noticed as an initial sign of Potomac horse


fever.
14 Bacterial diseases

11 12

11, 12 Ulcerative colitis. The colon is severely congested with prominent distended mesocolonic lymph vessels (11). The mucosa is
thickened and oedematous with multiple mucosal ulcerations and haemorrhages (12). These lesions may be indicative of Potomac
horse fever.

Diagnosis mucosal congestion and haemorrhages with


Using mice inoculation, N. risticii was first detected superficial epithelial erosive to necrotizing lesions
in the blood on post-inoculation day 10, peaked on and fibrin deposits. Furthermore, crypt abscesses and
post-inoculation day 19, and was not detectable after mixed inflammatory hypercellularity of the lamina
post-inoculation day 24. The N. risticii titre was propria may be present. Rickettsial clustered
maximal during the peak increase in rectal organisms of <1 μm can be identified in special silver
temperature, and infected horses seroconvert as stains within the apical cytoplasm of cryptal
detected by IFA assay and enzyme-linked enterocytes and in macrophages in the lamina
immunosorbent assay (ELISA) with antibody titres propria (Jubb et al. 2007).
between 1:160–1:640 and >1:5,000, respectively
(Dutta et al. 1988). Management/Treatment
For diagnosis, preference is given to culture The only effective treatment is the administration of
or PCR. PCR was successfully used to detect tetracycline in the early stages of the disease as there
the organism directly from the blood buffy coat cells is no truly effective vaccine available (Dutta et al.
of infected horses. It was estimated that buffy coat 1998, Rikihisa et al. 2004). IV administration of
cells obtained from less than 1 ml of blood from oxytetracycline (6.6 mg/kg BW sid for 5 days) is an
infected horses was adequate for the detection of N. effective treatment when given early in the clinical
risticii (Biswas et al. 1991). N. risticii was detected in course (Palmer et al. 1992). There is protective
the blood by nested PCR in 81% of the culture- immunity against N. risticii infection, as evidenced
positive clinical specimens, indicating that the nested by clinical resistence to reinfection for as long as 20
PCR is as sensitive as culture for detecting infection months after the initial infection (Palmer et al. 1990).
with N. risticii (Mott et al. 1997). However, Despite treatment with oxytetracycline (6.6 mg/kg
characteristic cytoplasmic inclusion bodies (either BW bid IV beginning 14 hours before inoculation
morulae or elementary bodies) can be visualized in and continuing for 10 days) before inoculation, the
the monocytes on a Wright–Giemsa- or H&E- antigenic stimulation was sufficient to induce such
stained smear of peripheral blood obtained during protective immunity (Palmer et al. 1988).
peak ehrlichiaemia (Holland et al. 1985).
Public health significance
Pathology Not convincing as yet.
Pathological features usually observed are mild
to moderate typhlitis and colitis, with congested and
ulcerated mucosae (11, 12), most prominent in the
right dorsal colon, and mesenteric lymphadenopathy.
Similar less grave lesions may be present in the
stomach and small intestine. Subcutaneous oedema
and laminitis may be accompanying gross features.
Microscopically, intestinal lesions are composed of
Bacterial diseases 15

Bartonella henselae: BARTONELLOSIS Public health significance


Phylum BXII Proteobacteria The bartonelloses of medical importance comprise
Class I Alphaproteobacteria/Order VI Carrión’s disease, trench fever, cat-scratch disease,
Rhizobiales/Family III Bartonellaceae/Genus I bacillary angiomatosis, and peliosis hepatis.
Bartonella The Bartonella spp. are considered emerging human
pathogens (Maguiña et al. 2009). B. henselae has
Definition/Overview been identified as the causative agent of cat-scratch
Bartonella spp. are associated with an extended disease. On the other hand B. quintana which causes
animal host range (including equines) and are the body lice-mediated trench fever in humans
identified as emerging in Europe (Vorou et al. 2007). and had no known animal reservoir, was shown to
infect a domestic cat (Vorou et al. 2007).
Aetiology Furthermore, Bartonella spp. were first recognized
Bartonella (formerly Rochalimaea species) spp. are to cause endocarditis in humans in 1993 when cases
members of the α-proteobacteria group that includes caused by B. quintana, B. elizabethae, and
the genera Rickettsia, Ehrlichia, Brucella, and the B. henselae were reported. Since the first isolation of
plant pathogen Agrobacterium tumefaciens. They B. vinsonii subsp. berkhoffii from a dog with
are fastidious, Gram-negative short-to-spirillar endocarditis, this organism has emerged as an
bacteria that occur in the blood of man and other important pathogen in dogs and an emerging
mammals; they are usually vector borne but can also pathogen in people (Chomel et al. 2009).
be transmitted by animal scratches and bites from
haematophagous insects, such as sandflies
(Lutzomyia spp.), fleas, and lice (Maguiña et al.
2009).

Incubation period
Not established in the equine species yet.

Clinical presentation
B. henselae was isolated from the blood of a horse
with chronic arthropathy and a horse with
presumptive vasculitis (Jones et al. 2008).

Diagnosis
Blood samples can be tested for the presence
of Bartonella spp. by a combination of multiplex
real-time PCR and enrichment culture technique
(Jones et al. 2008).

Pathology
B. henselae infection caused abortion of a foal
exhibiting necrosis and vasculitis in multiple tissues,
with intralesional Gram-negative short-to-spirillar
bacteria (Johnson et al. 2009).

Management/Treatment
Human Bartonella isolates are highly susceptible to
antibiotics, including most of the beta-lactams, the
aminoglycosides, the macrolides, doxycycline, and
rifampicin (rifampin) (Maurin et al. 1995).
16 Bacterial diseases

Brucella spp.: BRUCELLOSIS In horses admitted for evaluation of fistulous


Phylum BXVII Spirochaetes withers, 38% tested for antibody to B. abortus were
Class I Alphaproteobacteria/Order VI seropositive. Horses that tested seropositive were
Rhizobiales/Family IV Brucellaceae/Genus I significantly more likely to have been pastured with
Brucella: Gram-negative aerobic rods and cocci cattle that were seropositive for B. abortus, and were
also significantly more likely to have had
Definition/Overview radiographic evidence of vertebral osteomyelitis than
Coincidental infection in horses caused by various were horses that tested seronegative (Cohen et al.
bacteria of the genus Brucella is especially associated 1992).
with abortion and fistulous withers. Brucellosis has
important public health significance and is a Pathophysiology
reportable disease. These bacteria do not produce classical virulence
factors, and their capacity to survive and replicate
Aetiology successfully within a variety of host cells underlies
Brucellae are facultative intracellular, Gram-negative, their pathogenicity. Extensive replication of the
partially acid-fast coccobacilli. The bacterium is 0.5– Brucellae in placental trophoblasts is associated with
0.7 μm in diameter and 0.6-1.5 μm in length. They reproductive tract pathology in natural hosts, and
are oxidase, catalase, and urease positive. Brucella prolonged persistence in macrophages leads to the
species considered important agents of human chronic infections that are a hallmark of brucellosis
disease include B. melitensis, B. abortus, and B. suis in both natural hosts and humans (Roop et al. 2009).
(Ekers 1978, Mohandas et al. 2009). Isolation of B.
suis biotype 1 (Cook & Kingston 1988) and Incubation period
B. abortus biotypes 1, 2, and 4 (Ekers 1978, Experimental intraconjunctival infection of
Carrigan et al. 1987) was reported from horses. horses with B. abortus revealed no appreciable
clinical signs up to 30 months except mild
Epidemiology pyrexia (MacMillan et al. 1982, MacMillan &
The epidemiology of human brucellosis, the Cockrem 1986).
commonest zoonotic infection worldwide, has
drastically changed over the past decade because of Clinical presentation
various sanitary, socioeconomic, and political The clinical signs of the disease are variable, but
reasons, together with the evolution of international include fistulous withers (Cohen et al. 1992),
travel. Several areas traditionally considered to be abortion (Shortridge 1967, McCaughey &
endemic, such as France, Israel, and most of Latin Kerr 1967), arthritis (Carrigan et al. 1987), and
America, have achieved control of the disease. On vertebral osteomyelitis (Collins et al. 1971).
the other hand, new foci of human brucellosis have
emerged, particularly in central Asia, while the Differential diagnosis
situation in certain countries of the near East (e.g. The differential diagnosis predominantly includes
Syria) is rapidly worsening (Pappas et al. 2006). B. various causes of abortion and fever (see p. 263).
melitensis biovar 3 is the most commonly isolated
species from animals in Egypt, Jordan, Israel, Diagnosis
Tunisia, and Turkey (Refai 2002). The The diagnosis is based on a positive culture of the
seroprevalence of Brucella species among horses in bacterium and/or seroconversion as assessed by a
Jordan was 1% and 8.5% in donkeys. Contact with complement fixation test eventually combined with
small ruminant herds with a history of brucellosis a positive reaction to the intradermal skin test.
was associated with a high odds ratio (20 and 81 for However, an intradermal skin test was positive in
horses and donkeys, respectively) for Brucella infected adults only, and negative in all foals tested
seropositivity in equids (Abo-Shehada 2009). It has (MacMillan & Cockrem 1986).
been suggested that equines with a seroprevalence Antibodies to B. abortus became detectable from
rate of 0.2% are not a reservoir of brucellosis and the second week after inoculation. Titres in the
do not play an important role in the epidemiological serum agglutination and complement fixation tests
patterns of this disease in northeastern Mexico declined substantially after 6–8 weeks but reactions
(Acosta-González et al. 2006), with horses in Latin to the Coombs antiglobulin, 2-mercaptoethanol, and
America mainly infected with B. abortus and B. suis immunodiffusion tests were maintained (MacMillan
(Lucero et al. 2008). et al. 1982). Of interest, genus-specific real-time PCR
Bacterial diseases 17

assays, e.g. based on the bcsp31 gene, will lead to an brucellosis (Mazokopakis et al. 2003). Uveitis is also
early diagnosis but for the purpose of seen (Rolando et al. 2009). The standard treatment
epidemiological surveillance a species-specific real- for acute and chronic brucellosis is a combination of
time PCR deriving from the conventional AMOS doxycycline with a second drug such as rifampicin
(AbortusMelitensisOvisSuis)-PCR is necessary or gentamicin, in order to treat and to prevent
(Al Dahouk et al. 2004). complications and relapse (Sakran et al. 2006).

Pathology
Post-mortem examination of a foal suffering from
brucellosis disclosed granulomatous lesions in the
lungs, liver, testes, and metatarsophalangeal synovial
membranes. B. abortus identical with strain 544 was
recovered from lymphoid and other tissues
(MacMillan & Cockrem 1986).

Management/Treatment
Horses with a tentative diagnosis of brucellosis
should be isolated to prevent possible human
exposure. Treatment should not be attempted as the
pathogen has important public health significance
and brucellosis is a reportable disease. The most
commonly used veterinary vaccines are B. abortus
S19 and B. melitensis Rev.1 vaccines. B. abortus
RB51 vaccine is used in some countries on a small
scale. Vaccination is limited to cattle and small
ruminants (Refai 2002). Five horses that were
seropositive for B. abortus were administered strain
19 Brucella vaccine SC (n = 1) or IV (n = 4). The
horse treated by SC injection of vaccine improved
during hospitalization, but was lost to follow-up
evaluation. Three of four horses treated by
IV injection died, but one horse recovered within 4
weeks of treatment (Cohen et al. 1992).

Public health significance


Brucellosis has important public health significance.
Brucellosis, especially caused by B. melitensis
particularly biovar 3 (Refai 2002), remains one of
the most common zoonotic diseases worldwide with
more than 500,000 human cases reported annually
(Seleem et al. 2010). Involvement of the
musculoskeletal system is the most common
complication of human brucellosis, while
neurobrucellosis (like meningitis) and endocarditis
are life-threatening complications (Ranjbar et al.
2009).
Cardiovascular complications occur in <2%, but
account for most of the mortality. Brucella
endocarditis usually involves normal native aortic
valves in 75% of cases. A combination of antibiotics
and valve replacement is the most acceptable
treatment (Mohandas et al. 2009). Cutaneous
manifestations including erythema nodosum are not
specific and affect 1–14% of patients with
18 Bacterial diseases

Burkholderia mallei: ‘GLANDERS’ Clinical presentation


Phylum BXVII Spirochaetes Clinical signs include febrile episodes, cough, blood-
Class II Betaproteobacteria/Family encrusted material on nostrils, inflammatory nodules
Burkholderiaceae/Genus I Burkholderia: Gram- and ulcers developed in the nasal passages with a
negative aerobic rods and cocci sticky yellow discharge, characteristic stellate scars
in the nasal septum, purulent nasal discharge,
Definition/Overview enlargement of submaxillary lymph nodes, chronic
Glanders is an ancient, highly fatal, and usually lymphangitis, skin abscessation, progressive debility,
chronic respiratory disease of solipeds caused orchitis, and dyspnoea associated with interstitial
by Burkholderia mallei (formerly Pseudomonas pneumonia. Furthermore, apparent neurological
mallei) with humans being accidental hosts. The degeneration is seen in acute glanders (Lopez et al.
diagnosis is based on the presence of characteristic 2003). Life expectancy was judged likely to have
stellate scars in the nasal septum and a positive been less than 12 hours in B. mallei inoculated
reaction to the mallein test, combined with a positive horses due to subsequent pulmonary oedema (Lopez
culture of B. mallei. Human infections are often fatal et al. 2003).
if untreated.
Differential diagnosis
Aetiology The differential diagnosis includes various causes of
B. mallei is a facultative rod-shaped Gram-negative fever and dyspnoea (see p. 262).
nonspore-forming, nonmotile, intracellular pathogen
that can invade, survive, and replicate in epithelial Diagnosis
and phagocytic cell lines (Ribot & Ulrich 2006). It is The presence of stellate scars in the nasal septum is
an obligate animal pathogen whose natural hosts are regarded as pathognomonic. B. mallei can be
horses, donkeys, and mules, but infections can also cultured easily from purulent nasal discharge and the
occur in felines, camels, and goats. Virulence in complement fixation test can be used for serology.
B. mallei is multifactorial and several virulence Furthermore, the mallein test can also be used to
determinants have been identified and characterized identify infected horses; purulent exudate in the eye
(Schell et al. 2007). Seventeen distinct ribotypes were associated with blepharospasm of a glanderous
identified from human and equine infections (Harvey animal 24–48 hours following subconjunctival
& Minter 2005). inoculation is regarded as a positive test result.
Alternatively, the intracutaneous mallein test can be
Epidemiology used with an increase in rectal temperature and a
Glanders is endemic in Africa, Asia, the Middle East, swelling at the point of injection regarded as a
and Central and South America. Carriers that have positive test result (Arun et al. 1999). When tested
made an apparent recovery from the disease are the comparatively with Dutch PPD mallein as standard,
most important source of infection, as the pathogen trichloroacetic acid-precipitated proteins were
does not survive for more than 6 weeks outside the comparable to Dutch PPD mallein in potency and
host (Lehavi et al. 2002). innocuity, whereas ammonium sulphate-precipitated
proteins elicited nonspecific reactions (Verma et al.
Pathophysiology 1994).
Equines are generally infected orally (Schell et al. Competitive enzyme-linked immunosorbent assay
2007). Following penetration of the mucosae, the (cELISA) test specificity for B. mallei was 99%.
pathogen is spread via the lymphatic tissues. Concordance and kappa value between the
complement fixation (CF) and the cELISA
Incubation period procedures for the serodiagnosis of B. mallei
The incubation period varies from 1 to 2 days infection in experimentally exposed horses were
following intratracheal deposition, with rectal 70% and 0.44, respectively (Katz et al. 2000). The
temperatures increased to above 40°C (Lopez et al. cELISA offers the possibility for automatization, can
2003). be applied to noncomplement fixing sera, and used
for various host species although the complement
fixation test (CFT) is internationally mandatory for
testing of equine sera for the absence of glanders to
date (Sprague et al. 2009).
Bacterial diseases 19

13 Hypopyon, suppurative uveitis. The anterior eye chamber 13


is blurred with specks of a fibrinopurulent exudate. From
this foal Burkholderia cepacia was isolated. Members of the
B. cepacia complex are regarded as opportunistic pathogens.

Hydrolysis probe-based real-time PCR using the Public health significance


uneven distribution of type III secretion Humans are accidental hosts of B. mallei and the
system genes afforded considerable improvements majority of cases have been the result of
in the specificity and rapidity of the diagnosis of occupational contact with infected horses. Whereas
B. pseudomallei, B. mallei, and B. thailandensis, equines are generally infected orally, the primary
and allows rapid discrimination from opportunistic route of infection in humans is contamination of skin
pathogens such as members of the B. cepacia abrasions or mucous membranes with nasal
complex (13), that routine diagnostic laboratories discharge or skin lesion exudate from an infected
are more likely to encounter (Thibault et al. 2004). animal (Schell et al. 2007). Person-to-person spread
of B. mallei is extremely rare. In humans, glanders
Pathology is characterized by initial onset of fever, rigors and
B. mallei infection results in pyogranulomatous and malaise, culminating in a rapid onset of pneumonia,
necrotic pulmonary nodules, and ulcerative nodular bacteraemia, pustules and abscesses, leading to death
skin and respiratory mucosal lesions with in 7–10 days without antibiotic treatment. The
characteristic white stellate scars in the nasal septum. course of infection is dependent on the route of
Histologically, lung lesions comprise liquefactive exposure. Direct contact with the skin can lead to a
necrosis including neutrophils and surrounding localized cutaneous infection. Inhalation of aerosol
epithelioid macrophages and fibrosis. The dermal or dust containing B. mallei can lead to septicaemic,
disease of ulcerations including lymphangitis is pulmonary, or chronic infections of the muscle, liver,
named ‘farcy’ (Jubb et al. 2007). Remarkably, and spleen. The disease has a 95% case fatality rate
Streptococcus equi subsp. zooepidemicus was for untreated septicaemia infections and a 50% case
isolated from the brain of all B. mallei inoculated fatality rate in antibiotic-treated individuals
horses (Lopez et al. 2003). (Mandell et al. 1995). Burkholderia infections are
difficult to treat with antibiotics and no vaccine
Management/Treatment exists (Whitlock et al. 2007).
Horses with a tentative diagnosis of glanders should
be isolated to prevent possible human exposure.
Treatment should not be attempted as the pathogen
has important public health significance and
glanders is a reportable disease.
20 Bacterial diseases

Burkholderia pseudomallei: MELIOIDOSIS Incubation period


Phylum BXVII Spirochaetes This is not established in the equine species yet. The
Class II Betaproteobacteria/Family incubation period in man from defined inoculating
Burkholderiaceae/Genus I Burkholderia: Gram- events was previously ascertained as 1–21 (mean 9)
negative aerobic rods and cocci days (Currie et al. 2000b).

Definition/Overview Clinical presentation


Melioidosis is a rare disease caused by Burkholderia Clinical signs include fever, septicaemia, oedema,
pseudomallei (formerly Pseudomonas pseudomallei) colic, diarrhoea, and lymphangitis of the legs. A case
characterized by an intracellular life cycle. Both of acute meningoencephalomyelitis caused by
humans and animals (including birds, crocodiles, infection with B. pseudomallei has been described
and kangaroos) are susceptible to melioidosis with associated with inability to stand, opisthotonus,
both latency and a wide range of clinical facial paralysis (14) and nystagmus, rapidly
manifestations. Some species may develop progressing to violent struggling (Ladds et al. 1981).
melioidosis only if immunocompromised. Sheep,
goats, and horses are particularly susceptible, Differential diagnosis
but zoonotic transmission to humans is The differential diagnosis includes various causes of
extremely unusual (Neubauer et al. 1997, Choy et al. internal abscessation (without characteristic stellate
2000). Melioidosis has important public health scars in the nasal septum as seen in B. mallei) (see p.
significance and is a reportable disease. 262). Listeriosis should be considered in a case of
meningitis.
Aetiology
B. pseudomallei is a Gram-negative, bipolar-staining, Diagnosis
pleomorphic, motile bacillus, which is principally an B. pseudomallei can be cultured easily from purulent
environmental saprophyte responsible for nasal discharge. The diagnosis is based on a positive
melioidosis. reaction to the mallein test combined with a positive
culture.
Epidemiology
This saprophyte inhabitant of telluric environments
is mainly encountered in southeast Asia and northern
Australia, but is sporadically isolated in subtropical
and temperate countries (White 2003). Melioidosis
has become an increasingly important disease in
endemic areas such as northern Thailand and
Australia (Currie et al. 2000a). In endemic areas, the
positive rates of antibodies against B. pseudomallei
in humans, horses, oxen, and pigs were 4–15%, 9– 14
18%, 7–33%, and 35%, respectively (Li et al. 1994).

Pathophysiology
Following ingestion via contaminated soil or faeces,
a diverse assortment of virulence factors (quorum
sensing, type III secretion system, lipopolysaccharide
and other surface polysaccharides) allows B.
pseudomallei to become an effective opportunistic
pathogen; its intracellular life cycle also allows it to
avoid or subvert the host immune system (Adler et
al. 2009, Wiersinga & van der Poll 2009). The BoaA
and BoaB genes specify adhesins that mediate
adherence to epithelial cells of the human respiratory
tract. The BoaA gene product is shared by B.
pseudomallei and B. mallei, whereas BoaB appears
to be a B. pseudomallei-specific adherence factor
(Balder et al. 2010).

14 Facial paralysis is associated with equine melioidosis.


Bacterial diseases 21

Hydrolysis probe-based real-time PCR methods 15


using the uneven distribution of type III secretion
system genes afford considerable improvements
in the specificity and rapidity of the diagnosis of
B. pseudomallei, B. mallei, and B. thailandensis and
allow rapid discrimination from opportunistic
pathogens, such as members of the B. cepacia
complex (15, 16), that routine diagnostic labora-
tories are more likely to encounter (Thibault et al.
2004).

Pathology
Multiple abscesses in most organs are characteristic
of the disease. The encapsulated nodules with 15 Necrosuppurative bronchopneumonia in a foal. The
caseous centres are composed of necrosis, cranioventral lung field is hyperaemic, consolidated, and firm.
neutrophils, lymphocytes, and epithelioid macro- Lesions resemble pulmonary lesions in melioidosis. From this
phages. In a case of acute meningoencephalomyelitis foal Burkholderia cepacia was isolated.
gross examination revealed malacia and
haemorrhage in the medulla oblongata and adjacent
spinal cord. Microscopically there were disseminated
focal neutrophilic accumulations in affected areas, 16
perivascular cuffing with mononuclear cells and
lymphocytes, and marked oedema. Intracellular
bacteria were identified in sections stained by the
Giemsa method (Ladds et al. 1981).

Management/Treatment
Horses with a tentative diagnosis of melioidosis
should be isolated to prevent possible human
exposure. Treatment should not be attempted as the
disease has important public health significance.
Furthermore, the ubiquitous bacterium is
characterized by remarkable insensitivity to
antimicrobial drugs. For instance, B. pseudomallei is 16 Necrosuppurative bronchopneumonia in a foal. The
intrinsically resistant to aminoglycosides and cranioventral lung lobes show on cut section a well-delineated
macrolides, mostly due to AmrAB-OprA efflux hyperaemic area enclosing pale yellow, variably sized,
pump expression (Trunck et al. 2009). caseating, coagulative, necrosuppurative sequesters of
Immunization with heat-inactivated B. pseudo- remnant pulmonary parenchyma. Lesions resemble
mallei cells provided the highest levels of protection pulmonary lesions in melioidosis. From this foal Burkholderia
against either melioidosis or glanders, indicating cepacia was isolated.
longer-term potential for heat-inactivated bacteria
to be developed as vaccines against melioidosis and
glanders (Sarkar-Tyson et al. 2009).

Public health significance


Melioidosis has important public health significance
and is a reportable disease. It is a life-threatening
disease that is mainly acquired through skin
inoculation or pulmonary contamination, although
other routes have been documented (Neubauer et al.
1997). Primary skin melioidosis occurred in 12% of (Gibney et al. 2008). Severe septicaemia secondary
human patients. Secondary skin melioidosis to melioidosis carries a high mortality. Although
(multiple pustules from haematogenous spread) was melioidosis can involve most tissues and organs,
present in 2%. Patients with primary skin pericardial involvement is rare (De Keulenaer et al.
melioidosis were more likely to have chronic 2008). Of human cases, 46% were bacteraemic and
presentations (duration of a minimum of 2 months) 19% died (Currie et al. 2000a).
22 Bacterial diseases

Bordetella bronchiseptica Incubation period


Phylum BXVII Spirochaetes Not established in the equine species yet.
Class II Betaproteobacteria/Family
III Alcanigenaceae/Genus III Bordetella: Gram- Clinical presentation
negative aerobic rods and cocci Clinical presentation includes respiratory disease in
foals (17) (Koehne et al. 1981), coughing in
Definition/Overview Thoroughbred racehorses (Christley et al. 2001),
The opportunistic bacterium Bordetella bronchi- bronchopneumonia (Saxegaard et al. 1971),
septica is a rare cause of acute respiratory disease abortion (Mohan & Obwolo 1991), and infertility
and abortion/infertility. (Mather et al. 1973). B. parapertussis did not grow
in tracheobronchial washing from a horse (Porter &
Aetiology Wardlaw 1994).
Pasteurellaceae are Gram-negative bacteria with an
important role as primary or opportunistic, mainly Differential diagnosis
respiratory, pathogens in domestic and wild animals. The differential diagnosis includes various causes of
Some species of Pasteurellaceae cause severe diseases fever and dyspnoea (see p. 262).
with high economic losses in commercial animal
husbandry and are of great diagnostic concern Diagnosis
(Dousse et al. 2008). Sixteen distinct ribotypes were Diagnosis primarily depends on culture of the
identified in B. bronchiseptica strains (Register et al. bacterium from tracheobronchial washing samples
1997). Four main types of variation of the combined with clinical signs. Analysis of
B. bronchiseptica lipopolysaccharide (LPS) are tracheobronchial washing samples for known
apparent: (1) heterogeneity of the core, (2) presence Bordetella nutrients revealed concentrations of
or absence of 0-chains, (3) differences at the level of amino acids and nicotinic acid averaging 0.35 mM
the hinge region between the 0-chain and the core, and 0.56 μg/ml, respectively (Porter & Wardlaw
and (4) differences in the association with other cell 1994).
surface constituents. Isolates from different animal
species did not show significant differences in their Pathology
patterns of reactivity with monoclonal antibodies Common lesions caused by B. bronchiseptica include
(LeBlay et al. 1997). a catarrhal to suppurative bronchopneumonia and a
(sero)fibrinous pleuropneumonia. These are usually
Epidemiology opportunistic secondary infections preceded by viral
Glucose nonfermenting Gram-negative bacilli have infections in juvenile animals.
been recognized as opportunistic pathogens of
humans. The most common veterinary glucose Management/Treatment
nonfermenting Gram-negative bacilli were Treatment of diseased animals is supportive and
Pseudomonas aeruginosa, Acinetobacter calcoaceticus, specific treatment should be based on in-vitro
B. bronchiseptica, and Pseudomonas pseudoalcaligenes. antimicrobial susceptibility testing.
Of all clinical veterinary specimens submitted for
cultures, 10% contained nonfermenters (Mathewson Public health significance
& Simpson 1982). B. bronchiseptica was isolated The absence of smooth-type LPSs appears to be
from bronchial lavage specimens in distal respiratory rather frequent in human isolates, since long-chain
tract disease (nasal discharge, cough, pneumonia) in LPSs were detectable in only 52% of human isolates,
13% of foals (1–8 months old) (Hoffman et al. whereas 94% of animal isolates contained molecules
1993). of that type (Le Blay et al. 1997). B. bronchiseptica
might have some public health significance and its
Pathophysiology zoonotic risk should be minimized.
Either B. bronchiseptica does not persist inside
animals or susceptible animals possess specific
receptors for smooth-type LPSs, in contrast to man
(Le Blay et al. 1997).
Bacterial diseases 23

17

17 Suppurative bronchopneumonia in a foal. Cranioventral pulmonary hyperaemia and


consolidation. A primary viral respiratory infection (herpesvirus, influenza virus) can be
complicated by opportunistic bacteria like Streptococcus spp., Escherichia coli, Klebsiella
pneumoniae, Rhodococcus equi, and Bordetella bronchiseptica.
24 Bacterial diseases

Taylorella equigenitalis: CONTAGIOUS latest CEM occurrence was warranted (Timoney


EQUINE METRITIS 2011). Among stallions examined in Slovenia, 92%
Phylum BXVII Spirochaetes were negative to T. equigenitalis by either PCR or
Class II Betaproteobacteria/Family III culture (Zdovc et al. 2005). In comparison, from
Alcanigenaceae/Genus XI Taylorella: Gram- 1999 through 2001, four out of 120 imported
negative, microaerophilic, fastidious slow-growing European stallions tested positive for CEM at a
coccobacilli quarantine facility in Darlington, MD, USA (Kristula
& Smith 2004). Samples from mares with no clinical
Definition/Overview signs of CEM submitted for conventional culture
Contagious equine metritis (CEM) caused by were negative for T. equigenitalis, but in the PCR
Taylorella equigenitalis is a highly contagious disease assay 49% were positive for Taylorella DNA. The
that is transmitted venereally. The carrier state high incidence of Taylorella in horse populations
occurs in the mare and the stallion and carrier without apparent clinical signs of CEM, the
animals are frequently the source of infection for occurrence of incidental clinical cases, and the
new outbreaks (Timoney 1996). known variability between strains indicate that
Taylorella was endemic in the horse population
Aetiology (Parlevliet et al. 1997).
T. equigenitalis is a Gram-negative, microaerophilic,
fastidious slow-growing coccobacillus with Pathophysiology
streptomycin-sensitive and -resistant biotypes CEM is transmitted by direct or indirect venereal
(Timoney 1996). Isolates of T. equigenitalis obtained contact. The invasiveness of T. equigenitalis strains
from European horses analysed by pulsed-field gel seemed to be associated with the contagiousness of
electrophoresis (PFGE) were classified into 18 the infection, whereas the replication index seemed
genotypes (Kagawa et al. 2001). High sequence to be associated with the severity of the symptoms
similarity (99.5% or more) was observed throughout of contagious equine metritis (Bleumink-Pluym et al.
isolates from Japan, Australia, and France, except 1996).
from nucleotide positions 138 to 501 where
substitutions and deletions were noted (Matsuda et Incubation period
al. 2006). A phylogenetic analysis revealed a position Horses challenged with T. equigenitalis showed
of T. equigenitalis in the beta subclass of the class seroconversion from day 11 post-inoculation (Katz
Proteobacteria apart from the position of Haemo- & Geer 2001).
philus influenzae, which belongs in the gamma
subclass of Proteobacteria. A close phylogenetic Clinical presentation
relationship among T. equigenitalis, Alcaligenes CEM can be the cause of short-term infertility
xylosoxidans, and Bordetella bronchiseptica was sometimes associated with mucopurulent discharge
detected (Bleumink-Pluym et al. 1993). Lipo- and, very rarely, abortion in mares (Fontijne et al.
polysaccharide O-PS could be a specific marker for 1989). Unlike the mare, stallions exposed to T.
identification and differentiation of T. equigenitalis equigenitalis do not develop clinical signs of disease
and T. asinigenitalis, and provide the basis for the (Timoney 1996). It has been concluded that T.
development of specific detection assays for equigenitalis is of limited significance in horse
T. equigenitalis (Brooks et al. 2010). breeding (Parlevliet et al. 1997).

Epidemiology Differential diagnosis


CEM has given rise to international concern since it Atypical (donkey-origin) Taylorella spp. infections
was first recognized as a novel venereal disease of should be considered as a differential diagnosis
equids in 1977. The first known outbreak of CEM in of equine infertility in mares (Katz et al. 2000).
the USA was in Kentucky in 1978. For some time T. asinigenitalis, resembling T. equigenitalis, was
none of the subsequent outbreaks impacted recently isolated from the urethral fossa, urethra,
significantly on the horse industry. That changed and penile sheath of a 3-year-old stallion of the
dramatically in 2008, however, after the discovery of Ardennes breed when it was routinely tested for
some 1,005 exposed and carrier stallions and mares CEM. However, the colony appearance, the slow
in 48 states. Neither clinical evidence of CEM nor growth rate, and the results in the API ZYM test
decreased pregnancy rates were reportedly a feature differed slightly from those of T. equigenitalis.
in infected or exposed mares. In light of these Sequence analysis of 16S rRNA genes was shown to
findings, the question arose as to whether or not the be a reliable tool for differentiation of donkey-
considerable expense incurred in investigating the related T. asinigenitalis from T. equigenitalis, as well
Bacterial diseases 25

18 19

18, 19 Taylorella asinigenitalis resembling T. equigenitalis


might be isolated from the urethral fossa (arrow) in horses.

as for identification of these species. The cervical swabs of 84% (Wood et al. 2005).
T. asinigenitalis strain had a low minimum inhibitory There was close agreement between CFT and
concentration (MIC) of gentamicin (≤1 μg/ml) but a ELISA methodologies during the post-exposure
high MIC of streptomycin (>16 μg/ml) (Båverud et time period used to detect CEM serodiagnostically
al. 2006). in regulatory animal health testing programmes.
Unlike the CFT, which requires an overnight
Diagnosis incubation step, the ELISAs are more convenient
Diagnosis is based primarily on culture of and can be completed in 3 hours (Katz & Geer
the bacterium from its predilection sites in 2001).
the reproductive tract of the mare and the stallion
(18, 19) (Timoney 1996). However, the rate of Pathology
T. equigenitalis detection was higher with PCR than Macroscopically no vaginal lesions are apparent; the
with the classic bacteriological examination. PCR is endometrial mucosa may be swollen and corrugated
especially valuable in cases of intensive bacterial and with a scant mucopurulent exudate. Histology of
fungal contamination of swabs where the isolation uterine biopsies might reveal a mild endometritis,
of T. equigenitalis usually fails (Zdovc et al. 2005). A characterized by interstitial mucosal oedema and a
direct-PCR assay was developed for the rapid mild inflammatory infiltrate composed of
detection of T. equigenitalis in equine genital swabs neutrophils; later plasma cells may be more evident
without need for a preliminary step of DNA (Jubb et al. 2007).
extraction or bacterial isolation (Duquesne et al.
2007). The assay is also able to discriminate between Management/Treatment
T. equigenitalis and T. asinigenitalis (Wakeley et al. Aggressive systemic antibiotic therapy accompanied
2006). by routine topical therapy might be required to treat
In chronically infected mares, the organism was CEM-positive stallions (Kristula & Smith 2004).
detectable in the clitoral swabs of nearly 93%, but
in the cervical swabs of only 31%. In contrast, in Public health significance
acutely infected mares, the organism was detectable Not convincing yet.
in the clitoral swabs of nearly 69%, but in the
26 Bacterial diseases

Francisella tularensis: TULAREMIA Pathology


Phylum BXII Proteobacteria Not established in the equine species yet.
Class III Gammaproteobacteria/Order
V Thiotrichales/Family II Francisellaceae/Genus I Public health significance
Francisella: Gram-negative aerobic rods and cocci Tularemia is regarded as an important (tickborne)
zoonosis with two primary disease manifestations,
Definition/Overview ulceroglandular and glandular. Two subspecies of F.
Tularemia caused by Francisella tularensis (formerly tularensis cause most human illness, namely
Pasteurella tularensis) is identified as emerging in subspecies tularensis, also known as type A, and
Europe (Vorou et al. 2007) although the subspecies holarctica, referred to as type B. The
pathogenicity of F. tularensis for the horse appears equine species is not regarded as a main reservoir for
to be extremely low. human infection in contrast with rodents and
lagomorphs (Petersen et al. 2009).
Aetiology
F. tularensis is a Gram-negative arthropod-borne
coccobacillus (Petersen et al. 2009).

Epidemiology
The serological response in burros and horses to the
viable LVS strain of F. tularensis generated high-
titred agglutinating antisera and fluorescent antibody
conjugates in both groups of animals. Maximum
titres were obtained in horses 14–21 days (up to
1:1,024 and 1:360, respectively) and in burros 21–28
days (up to 1:1,024 and 1:160, respectively) after the
start of vaccination. The use of so-called
Woodhour’s adjuvants or booster inoculations did
not result in increased titres (Green et al. 1970).

Pathophysiology
Free-living amoebae feed on bacteria, fungi, and
algae. However, some microorganisms have evolved
to become resistant to these protists. These amoeba-
resistant microorganisms include established
pathogens, such as F. tularensis, Legionella spp.,
Chlamydophila pneumoniae, and Listeria mono-
cytogenes. Free-living amoebae represent an
important reservoir of amoeba-resistant micro-
organisms and may, while encysted, protect the
internalized bacteria from chlorine and other
biocides. On the other hand, free-living amoebae
may act as a ‘Trojan horse’, bringing hidden
amoeba-resistant microorganisms within the human
or animal ‘Troy’, and may produce vesicles filled
with amoeba-resistant microorganisms, increasing
their transmission potential (Greub & Raoult 2004).

Incubation period
Not established in the equine species yet.

Clinical presentation
Not established in the equine species yet.
Bacterial diseases 27

Legionella pneumophila Clinical presentation


Phylum BXII Proteobacteria Signs of clinical illness were restricted to a transient
Class III Gammaproteobacteria/Order VI febrile response and lymphadenopathy (Cho et al.
Legionellales/Family I Legionellaceae/Genus I 1983).
Legionella: Gram-negative aerobic rods and cocci
Diagnosis
Definition/Overview Agglutinating antibodies persisted at least 4 months
Febrile lymphadenopathy can be experimentally after infection (Cho et al. 1983) with a high
induced by Legionella pneumophila. It has been correlation (r = 0.89) found between titres measured
concluded that there is no evidence to support a role by either the indirect fluorescent antibody test or the
for the horse in the maintenance of these organisms microagglutination test (Cho et al. 1984). All horses
in nature (Cho et al. 1983). exhibited a marked increase in agglutinating
antibodies to L. pneumophila serogroups 1 and 3 as
Aetiology early as 4 days after experimental challenge (Cho et
The pathogenicity of L. pneumophila serogroups 1 al. 1983).
and 3 for the horse appears to be low (Cho et al.
1983). Pathology
At necropsy, only moderate generalized
Epidemiology lymphadenopathy was noted with lymph nodes
Seroconversions in horses provided additional showing evidence of reactive hyperplasia.
evidence that horses become naturally exposed to Histologically, the lungs contained evidence of a low-
Legionella spp. Nineteen percent of horses grade inflammatory response characterized by focal
seroconverted to at least one serogroup (out of 4) of proliferation of alveolar lining cells, with few
L. pneumophila (Cho et al. 1984). With 58% of the neutrophils and eosinophils (Cho et al. 1983).
sera tested negative, 35% had end-point titres of 1:2,
7% end-point of 1:16 and 0.3% an end-point of Management/Treatment
1:256. South African serological results revealed a Not appropriate yet.
much lower exposure rate than that reported in the
USA (Wilkins & Bergh 1988). In addition, a high Public health significance
percentage of seropositivity suggested that horses are Not convincing yet as it has been stated that the
commonly infected with L. pneumophila or related horse could not be considered to be a source of
organisms, and the age-specific rates of occurrence infection but that both humans and animals were
indicated that infection was related directly to probably exposed to a common source of infection.
duration of exposure. The occurrence of positive Serological testing of people closely associated with
(1:64) equine sera (31%) was significantly higher horses showed that out of 22 people, three had a
than the occurrence of positive sera in cattle (5%), positive end-point titre of 1:64 and only one person
swine (3%), sheep (2%), dogs (2%), goats (0.5%), showed an end-point titre of 1:256 (Wilkins & Bergh
wildlife (0%), and humans (0.4%) as assessed by 1988).
means of microagglutination. The highest titre
measured in horses was 1:512. Of the positive sera in
horses, 44% reacted to a single serogroup (III or I
most commonly), and 56% reacted to multiple
serogroups (II and III or I, II, and III most
commonly) (Collins et al. 1982).

Pathophysiology
Not established in the equine species yet.

Incubation period
A transient decrease in circulating lymphocytes
occurred 2 days after inoculation (Cho et al. 1983).
28 Bacterial diseases

Coxiella burnetii: Q FEVER Clinical presentation


Phylum BXII Proteobacteria Moraxella spp. are associated with lymphoid
Class III Gammaproteobacteria/Order VI follicular hyperplasia (21) (Hoquet et al. 1985) and
Legionellales/Family II Coxiellaceae/Genus I conjunctivitis (Hughes & Pugh 1970, Huntington et
Coxiella al. 1987), although their clinical significance remains
unclear in the equine species.
Definition/Overview
Coxiella burnetii, the causative agent of Q fever, Diagnosis
is not currently reported to affect horses. Only Diagnosis primarily depends on culture of the
seropositivity was mentioned in horses ranging from bacterium in diseased animals combined with clinical
5.5–21.7% in Uruguay (Somma-Moreira et al. signs compared to negative controls.
1987).
Pathology
Moraxella spp. Moraxella spp. were isolated in 88% of horses with
Phylum BXII Proteobacteria pharyngitis of grades III and IV, followed
Class III Gammaproteobacteria/Order IX by Streptococcus equi subsp. zooepidemicus,
Pseudomonadales/Family II Moraxellaceae/Genus I Pseudomonas aeruginosa, coagulase-negative
Moraxella: Gram-negative aerobic rods and cocci staphylococci, and Enterobacter spp. (Hoquet et al.
1985).
Definition/Overview
Moraxella spp. are a frequent isolate in ocular and Management/Treatment
pharyngeal flora of clinically normal horses and Treatment of diseased animals is supportive.
horses suffering from lymphoid follicular hyperplasia
and conjunctivitis. Public health significance
Not convincing yet. Moraxella catarrhalis is an
Aetiology exclusively human pathogen and is a common cause
A Gram-negative, aerobic, oxidase-positive of otitis media in infants and children, causing 15–
diplococcus, that may colonize the conjunctiva and 20% of acute otitis media episodes. M. catarrhalis
the pharynx in horses. In ocular flora of clinically causes an estimated 2–4 million exacerbations of
normal horses (20), Corynebacterium spp., chronic obstructive pulmonary disease in adults
Staphylococcus spp., Bacillus spp., and Moraxella annually in the USA. Most strains produce beta-
spp. are the bacteria most frequently isolated lactamase and are thus resistant to ampicillin but
(Andrew et al. 2003), with Moraxella spp. susceptible to several classes of oral antimicrobial
comprising 28% of Gram-negative bacteria involved agents (Murphy & Parameswaran 2009).
(Gemensky-Metzler et al. 2005).

Epidemiology
There were no significant differences between the
number or type of organisms cultured during the
sampling seasons in ocular flora of clinically normal
Florida horses, whereas the likelihood of detecting
an organism depended on the horse’s age (Andrew
et al. 2003).

Pathophysiology
Unknown in the equine species yet.

Incubation period
Not established in the equine species yet.
Another random document with
no related content on Scribd:
5. Stylus, et Stigma, lente aucta.

SPECIFIC CHARACTER.

Heath, whose tips are bearded, and just within the blossom; the leaves grow
by fours, the flowers mostly by threes, and are smooth, cylindrically club-
shaped, an inch long, and of a deep blood colour.

DESCRIPTION.

Stem shrubby, grows two feet high, and upright; the branches grow
upright, having many smaller branches, which are thick set, and very short.
The Leaves grow by fours, are linear, smooth, shining, and of a deep
green, having very short foot-stalks, pressed to the stem.
The Flowers terminate the shorter branches near the summit of the
stem, forming as it were a long bunch; the foot-stalks are long, having three
floral leaves at their base.
Empalement. Cup four-leaved, which are lance-shaped, sawed, and
pressed to the blossom.
The Blossom is cylindrically club-shaped, smooth, blood colour, and an
inch long; the segments of the mouth are obtuse, and straight.
Chives. Eight hair-like threads, fixed into the receptacle; the tips are
bearded, and just within the blossom.
Pointal. Seed-vessel nearly egg-shaped, furrowed, and downy; Shaft
without the blossom; Summit four-cornered.
Native of the Cape of Good Hope.
Flowers from August, till December.

REFERENCE.

1. Empalement, and Blossom.


2. The Empalement magnified.
3. The Chives, and Pointal.
4. The Chives detached from the Pointal; one tip magnified.
5. The Shaft, and its Summit magnified.
ERICA cubica.

CHARACTER SPECIFICUS.

Erica, antheris basi bicornibus, sub-inclusis; stylus longissimus; corollis


sub-campanulatis, purureis, oris laciniis patulis, maximis, ovatis; foliis sub-
quaternis, obtusis.

DESCRIPTIO.

Caulis erectus, teres, ramosus, fruticosus, pedalis; rami et ramuli


filiformes, laxi, erecti.
Folia quaterna, sæpe quina, obtusa, reflexa, apice incurvata, glabra,
nitida, subtus sulcata; petiolis rubris, adpressis.
Flores sub-terminales, umbellati, cernui; pedunculi longissimi, angulati,
purpurei, foliolis binis, coloratis instructi.
Calyx. Perianthium tetraphyllum, foliolis ovatis, acuminatis, adpressis,
apicibus viridis.
Corolla globoso-campanulata, purpurea, limbo maximo, expanso;
laciniis ovatis, concavis.
Stamina. Filamenta octo capillaria, longitudine tubi, receptaculo inserta.
Antheræ bifidæ, ad basin bicornutæ.
Pistillum. Germen sub-globosum. Stylus filiformis, exsertus, corolla
duplo longior, purpureus. Stigma concavum, marginatum.
Habitat ad Caput Bonæ Spei.
Floret a mensi Aprili, in Julium.

REFERENTIA.

1. Calyx, et Corolla.
2. Calyx lente auctus.
3. Stamina, et Pistillum.
4. Stamina a Pistillo diducta; anthera una lente aucta.
5. Stylus, et Stigma, lente aucta.

SPECIFIC CHARACTER.

Heath, with tips two-horned at the base, nearly within the blossoms; shaft
very long; blossoms almost bell-shaped, and purple, having the segments of
the mouth spreading, very large, and egg-shaped; leaves growing mostly by
fours, and blunt ended.

DESCRIPTION.

Stem upright, cylindrical, branching, shrubby, growing a foot high; the


larger and smaller branches are thread-shaped, weak, and upright.
Leaves grow by fours, often by fives, blunt ended, bent back, and turned
inward at the point; smooth, shining, and channelled beneath; foot-stalks
red, and pressed to the stem.
The Flowers grow near the summits of the branches, in bunches,
hanging down; having very long foot-stalks, angled, purple, and two small,
coloured leaves on them.
Empalement. Cup four-leaved, which are egg-shaped, tapered, and
pressed to the blossom, the points green.
Blossom globularly bell-shaped and purple, the border very large and
spreading; the segments egg-shaped and concave.
Chives. Eight hair-like threads, the length of the tube, fixed into the
receptacle. Tips two-cleft, and two-horned at the base.
Pointal. Seed-bud nearly globular. Shaft thread-shaped, without the
blossom, twice its length, and purple. Summit concave, and bordered.
Native of the Cape of Good Hope.
Flowers from April, till July.

REFERENCE.
1. The Empalement, and Blossom.
2. The Empalement magnified.
3. The Chives, and Pointal.
4. The Chives detached from the Pointal; one tip magnified.
5. The Shaft, and Summit, magnified.
ERICA curviflora.

CHARACTER SPECIFICUS.

Erica, antheris muticis, sub-exsertis; corollis curvatis, clavato cylindricis,


pubescentibus, terminalibus, luteo-aurantiis; foliis quaternis, linearibus,
glabris.

DESCRIPTIO.

Caulis fruticosus, bipedalis; laxus, superne villosus; rami laxi, ramulosi,


ramulis brevissimis, frequentissimis, sparsis.
Folia quaterna, linearia, obtusa, glabra, subtus sulcata.
Flores terminales in ramulis, patenti, racemum longum formantes;
pedunculi brevissimi, foliolis tribus, adpressis.
Calyx. Perianthium tetraphyllum, foliolis subulatis, acuminatis, glabris,
adpressis.
Corolla curvata, clavata, pollicaris, pubescentia, luteo-aurantia; oris
laciniis expansis.
Stamina. Filamenta octo capillaria, curvata, longitudine tubi. Antheræ
muticæ.
Pistillum. Germen clavatum, sulcatum. Stylus filiformis, curvatus,
exsertus. Stigma tetragonum.
Habitat ad Caput Bonæ Spei.
Floret a mensi Julii, in Novembrem.

REFERENTIA.

1. Calyx, et Corolla.
2. Calyx, lente auctus.
3. Stamina, et Pistillum.
4. Stamina a Pistillo diducta; anthera una lente aucta.
5. Stylus, et Stigma, lente aucta.

SPECIFIC CHARACTER.

Heath, with beardless tips, just without the blossoms, which are curved,
cylindrically club-shaped, downy, terminating the branches, and of a yellow
gold colour; the leaves grow by fours, are linear, and smooth.

DESCRIPTION.

Stem shrubby, grows two feet high, weak, and hairy at the top; branches
weak and numerous; the smaller branches are very short, numerous, and
scattered.
Leaves grow by fours, are linear, blunt, smooth, and furrowed
underneath.
Flowers terminate the smaller branches, spreading out, and forming a
long spike; foot-stalks very short, with three small leaves pressed to the cup.
Empalement. Cup four-leaved, which are awl-shaped, tapering, smooth,
and pressed to the blossom.
Blossom curved, club-shaped, an inch long, downy, and of a yellow gold
colour; the segments of the mouth spread outward.
Chives. Eight hair-like threads, which are curved, and the length of the
blossom. Tips beardless.
Pointal. Seed-bud club-shaped, and furrowed. Shaft thread-shaped,
curved, and without the blossom. Summit four-cornered.
Native of the Cape of Good Hope.
Flowers from July, till November.

REFERENCE.

1. The Empalement, and Blossom.


2. The Empalement, magnified.
3. The Chives, and Pointal.
4. The Chives detached from the Pointal; one tip magnified.
5. The Shaft, and its Summit, magnified.
ERICA discolor.

CHARACTER SPECIFICUS.

Erica, antheris aristatis, inclusis; stylo exserto; corollis tubulosis,


subcylindraceis; foliis ternis.

DESCRIPTIO.

Caulis fruticosus, tripedalis, teres, erectus, gracilis, superne pubescens,


ramulis subsimplicibus.
Folia terna, linearia, erecto-patentia, supra plana, subtus revoluta, nitida,
saturate viridia.
Flores plerumque tres, subcernui, ramei, terminales, prope caulis
summitatem.
Calyx. Perianthium duplex, interius tetraphyllum, foliolis ovatis, erectis,
luteo-virentibus: exterius triphyllum, foliolis brevioribus priori
incumbentibus.
Corolla tubulosa, subcylindracea, ima parte, carnea, pallida, summa,
flavo-virescente, limbo quadrilobo, æquali erectiusculo.
Stamina. Filamenta octo capillaria, receptaculo inserta. Antheræ
oblongiusculæ, bipartitæ, inclusæ, aristatæ.
Pistillum. Germen oblongum. Stylus filiformis, corolla longior, apice
inflexo. Stigma subtetragonum, virescens.
Habitat ad Caput Bonæ Spei.
Floret a Novembri in Aprilem.

REFERENTIA.

1. Calyx, et Corolla.
2. Calyx lente auctus.
3. Stamina, et Pistillum.
4. Stamina a Pistillo diducta; anthera una lente aucta.
5. Stylus, et Stigma lente aucta.

SPECIFIC CHARACTER.

Heath, with bearded tips within the blossom; the pointal standing out; the
blossom tubular, nearly cylindrical; leaves growing by threes.

DESCRIPTION.

Stem shrubby, grows to three feet high, cylindrical, upright, slender,


downy at the top, the branches seldom divided.
Leaves growing by threes, linear, between upright and spreading, smooth
on the upper, and rolled back on the under, surface, shining and dark green.
Flowers are commonly three together, bending a little downward,
terminating the branches near the upper part of the stem.
Empalement. Cup double, the inner four-leaved, ovate, erect, and of a
greenish yellow: the outer three-leaved, the leaves shorter than the former,
and lying on them.
Blossom tubular, nearly cylindrical, of a light flesh colour at the base,
and yellowish green at the mouth, which is divided into four equal nearly
erect segments.
Chives. Eight hair-like threads, fixed into the receptacle. Tips nearly
oblong, divided, bearded, and within the blossom.
Pointal. Seed-vessel oblong. Shaft thread-shaped, longer than the
blossom, the point bending. The Summit nearly four-cornered, and greenish.
Native of the Cape of Good Hope.
Time of flowering from November till April.

REFERENCE.

1. The Empalement with the Blossom.


2. The Empalement magnified.
3. The Chives and Pointal.
4. The Chives detached from the Pointal, one tip magnified.
5. The Shaft, and its Summit magnified.
ERICA droseroides.

CHARACTER SPECIFICUS.

Erica, antheris cristatis, inclusis; corolla ventricosa, ore obliqua, purpurea;


pedunculis coloratis, longissimis; foliis alternis, sparsis, obtusis, pilis
glandulosis micantibus hirtis.

DESCRIPTIO.

Caulis erectus, spithameus; rami et ramuli filiformes, sparsi, erecto-


patentes, virgati.
Folia alterna, sparsa, linearia, obtusa, apice reflexa, pilis glandulosis
hirta.
Flores terminales, sub-umbellati, sub-erecti; pedunculi filiformes, foliis
duplo longiores; bracteæ minutæ, coloratæ.
Calyx. Perianthium tetraphyllum; foliolis sub-ovatis, viscosis,
acuminatis, coloratis, apicibus reflexis.
Corolla ventricosa, purpurea, costata, pubescens, ore obliqua, arctata,
profunde sanguinea; laciniis cordatis, expansis.
Stamina. Filamenta octo capillaria, torta, receptaculo inserta; antheræ
cristatæ, inclusæ.
Pistillum. Germen clavatum, sulcatum. Stylus filiformis, purpureus.
Stigma tetragonum.
Habitat ad Caput Bonæ Spei.
Floret a mense Julii ad Octobrem.

REFERENTIA.

1. Calyx et Corolla.
2. Calyx lente auctus.
3. Stamina et Pistillum.
4. Stamina a Pistillo diducta; antherâ unâ lente auctâ.
5. Stylus et Stigma lente aucta.

SPECIFIC CHARACTER.

Heath, with crested tips, within the blossom, which is big-bellied, oblique-
mouthed, and purple; the foot-stalks are coloured, and very long; leaves
grow alternate, scattered, blunt, and thick with shining glandular hairs.

DESCRIPTION.

Stem upright, about a span high; the larger and smaller branches are
thread-shaped, scattered, upright, spreading, and twiggy.
Leaves grow alternate, scattered, linear, blunt, the end bent back, and
thick with glandular hairs.
Flowers are terminal, nearly in bunches, almost upright; the foot-stalks
are thread-shaped, and twice as long as the leaves; the floral leaves are
small, and coloured.
Empalement. Cup four-leaved; leaves nearly egg-shaped, clammy,
pointed, coloured, and bent outward at the top.
Blossom big-bellied, purple, ribbed, and downy, with the mouth oblique,
narrowed, and of a deep blood colour; the segments are heart-shaped, and
spreading.
Chives. Eight hair-like threads, which are twisted, and fixed into the
receptacle; the tips are crested, and within the blossom.
Pointal. Seed-vessel club-shaped, and furrowed. Shaft thread-shaped,
and purple. Summit four-cornered.
Native of the Cape of Good Hope.
Flowers from the month of July till October.

REFERENCE.
1. The Empalement and Blossom.
2. The Empalement magnified.
3. The Chives and Pointal.
4. The Chives detached from the Pointal; one tip magnified.
5. The Shaft and its Summit magnified.
ERICA exsurgens.

CHARACTER SPECIFICUS.

Erica, antheris muticis, exsertis; foliis quaternis, mucronatis, apicibus


reflexis; corollis clavato-cylindraceis, verticillatis, aurantiis.

DESCRIPTIO.

Caulis fruticosus, pedalis et ultra, erectus, basi simplicissimus; rami


verticillati, simplices, erecti.
Folia quaterna, linearia, glabra, apice reflexa, mucronata, supra plana,
subtus sulcata, unguicularia, petiolis adpressis.
Flores in medio ramorum verticillati, recti; verticilli alter alteri
exsurgentes; pedunculi longi, bracteis tribus instructi.
Calyx. Perianthium tetraphyllum, foliolis lanceolatis, acuminatis,
carinatis, viscosis, adpressis.
Corolla clavato-cylindrica, pollicaria, aurantia; ore limbo quadrilobo
revoluto, laciniis superioribus longioribus.
Stamina. Filamenta octo capillaria, receptaculo inserta. Antheræ muticæ,
magnæ, exsertæ.
Pistillum. Germen clavatum, sulcatum. Stylus filiformis, staminibus
longior. Stigma tetragonum.
Habitat ad Caput Bonæ Spei.
Floret a mense Septembris, in Martium.

REFERENTIA.

1. Calyx, et Corolla.
2. Calyx lente auctus.
3. Stamina et Pistillum.

You might also like