Anda di halaman 1dari 81

Leptospirosis

Laboratory Manual

Regional Medical Research Centre


Indian Council of Medical Research
Port Blair
Leptospirosis

Laboratory Manual

Regional Medical Research Centre


Indian Council of Medical Research
Port Blair
Copyright World Health Organization (2007)

This document is not a formal publication of the World Health Organization (WHO) and all
rights are reserved by the Organization. The document may however be freely reviewed,
abstracted, reproduced or translated, in part or whole but not for sale or use in conjunction
with commercial purposes.

The views expressed in documents by named authors are solely the responsibility of those authors.

Design, layout and printing by:


New Concept Information Systems Pvt. Ltd., New Delhi.
Email: nc.communication@gmail.com
vkpk;Z ,u-ds- xkaxqyh Hkkjrh; vk;q f oZ K ku vuq l a / kku ifj"kn
egkfunskd oh- jkefya x Lokeh Hkou] va l kjh uxj]
iks L V ckW D l 4911] ubZ fnYyh & 110 029
MD, D.Sc (hc) FMedSci (London, FRC Path. (London), FAMS, Indian Council of Medical Research
FNA, FASc, FNASc, FTWAS (Italy), FIACS (Canada), FIMSA
V. Ramalingaswami Bhawan, Ansari Nagar,
Director General Post Box 4911, New Delhi - 110 029

Foreword
Leptospirosis is an important public health problem of India. Recently there has
been frequent resurgence of this malady coinciding with the natural calamity.
In spite of considerable advances in the management of this condition, timely
and accurate diagnosis is often difcult, owing to its atypical presentation and
similarity of signs and symptoms with other infectious diseases.

It is indeed a matter of great satisfaction to learn that RMRC, Port Blair in


collaboration with WHO is bringing out the much needed manual on laboratory
diagnosis of leptospirosis. This manual is a crystallization of more than a decade
of experiences and endeavour of the scientists of RMRC Port Blair working in
the eld of leptospirosis. This is a meticulously planned manual presented in a
simple way for its use in the laboratory for the diagnosis and characterization of
leptospirosis. This book will be of signicant utility and will prove to be a ready
reckoner for the clinicians and the laboratory personnel as well.

I sincerely compliment the joint efforts of RMRC, Port Blair and WHO in bringing
out this manual which I am sure will serve as a useful guide for early detection
of the disease, and thus reducing the morbidity and mortality associated with
leptospirosis.

(N.K. Ganguly)
Director General

Tele: 26588204 (Off.); PABX: 26589334, 26589335, 26589336, 26588707 Extn. : 264 (Res.): 26493145, 26493045
Fax: 91 - 11 - 26588662, 26589492, 26589647, 26589258; E-mail: gangulynk@icmr.org.in; gangulynk41@yahoo.co.in (personal)

Laboratory Manual iii


Preface

Leptospirosis is becoming an increasingly signicant public health problem, particularly in tropical


developing countries. The whole of Southeast and South Asia are endemic to the disease. Frequent
outbreaks are occurring, many of which in the aftermath of natural disasters. Yearly upsurges and
outbreaks are common in rice cultivating regions as a large number of farmers get exposed to
contaminated wet environment. Complications such as severe pulmonary haemorrhages and renal
failure are being reported more frequently. Once these complications set in, it is difcult to save
the patient even in most well-equipped hospitals and the case fatality ratio becomes very high.

In most of the developing countries where leptospirosis is endemic, no specic control programme
is in operation and the surveillance is often incomplete. Therefore, the disease outbreaks continue
unchecked and even an estimate of the disease burden is missing. Being a zoonotic disease with
a large spectrum of animal carriers and the difculty in preventing exposure of the people,
whose subsistence depends upon small scale farming and other occupations closely linked to the
environment, it is difcult to devise an effective control strategy. In this situation, early case
detection and treatment becomes very important for reducing the morbidity and mortality.

The two obstacles for early case detection are the lack of awareness of the people and medical
professionals about the disease and the unavailability of laboratory support for diagnosis. Because
of the frequent occurrence of the disease either in the form of outbreaks or as sporadic cases,
awareness, at least among the medical professionals is increasing. However, lack of laboratory
support and trained laboratory manpower is still an important issue in leptospirosis surveillance and
control. Several rapid test kits has become available in the market in the recent years. However,
there is no uniform standard or algorithm for laboratory diagnosis. There is a need to systematically
evaluate these commercially available tests and evolve a diagnostic algorithm.

This Centre has been carrying out research on leptospirosis for about one and a half decades now.
Since 1999 it is working as the National Leptospirosis Reference Centre and since 2003 as WHO
Collaborating Centre for Diagnosis, Research, Reference and Training in Leptospirosis. As part of
these activities, the Centre has also been conducting Hands-on-Training Workshop on laboratory
diagnosis of leptospirosis on alternate years.

We at this Centre thought it worthwhile to bring out a document based on the information
generated by the research activities of the Centre and the Collaborative efforts of this Centre
and WHO. This manual is a result of this. The primary objective is to briey present the
existing knowledge about the disease and its pathogen and draw guidelines for procedures for
laboratory diagnosis and characterization of leptospires. I hope this document jointly published
by the Regional Medical Research Centre (ICMR), Port Blair and WHO will address some of the
current issues in leptospirosis diagnosis, surveillance and control.

P. Vijayachari

Laboratory Manual v
Content
List of Abbreviations viii
CHAPTER 1
Introduction 1
CHAPTER 2
Historical Perspective 4
CHAPTER 3
Leptospira 8
CHAPTER 4
Epidemiology of Leptospirosis 13
CHAPTER 5
Clinical Manifestations 22
CHAPTER 6
Laboratory Diagnosis 27
CHAPTER 7
Serological Characterization of Leptospires 46
CHAPTER 8
Molecular Tools in the Diagnosis and Characterization of Leptospires 52
ANNEXURES
I. Preparation of EMJH Medium 61
II. Preparation of Fletchers Medium 62
III. Maintenance of Leptospires 63
IV. Purication of Contaminated Cultures 64
V. Preparation of Antisera 65
VI. Requirements for a Leptospirosis Laboratory 66
VII. Measurement of the Density of Leptospira Culture 67
VIII. General Safety Rules for Leptospirosis Laboratories 68
IX. Typing of Leptospira Isolates Report Format 69

Laboratory Manual vii


List of Abbreviations

5-FU - 5-Fluoro Uracil

AFLP - Amplied Fragment Length Polymorphism

Ag - Antigen

APPCR - Arbitrarily Primed PCR

CAAT - Cross Agglutination Absorption Test

DGM - Dark Ground Microscopy

DNA - Deoxyribonucleic Acid

EIA - Enzyme Immuno Assay

ELISA - Enzyme-Linked Immuno Sorbent Assay

EMJH - Ellinghausen McCullough Johnson Harris Medium

FAFLP - Fluorescent Amplied Fragment Length Polymorphism

IFA - Immuno Fluorescent Antibody Test

IHA - Indirect Haemagglutination Test

ILS - International Leptospirosis Society

Lepto-LAT - Latex Agglutination Test

mAbs - Monoclonal Antibodies

MAT - Microscopic Agglutination Test

MCAT - Microcapsule Agglutination Test

MSAT - Macroscopic Slide Agglutination Test

PCR - Polymerase Chain Reaction

PFGE - Pulsed Field Gel Electrophoresis

RAPD - Random Amplied Polymorphic DNA ngerprinting

REA - Restriction Enzyme nuclease Analysis

RFLP - Restriction Fragment Length Polymorphism

viii Leptospirosis
Chapter 1

Introduction
P. VIJAYACHARI

L
eptospirosis is an acute bacterial infection form is that presenting with severe pulmonary
caused by spirochetes belonging to the haemorrhage4, 10, 11. Other complications include
genus Leptospira1 that can lead to multiple acute respiratory failure14, myocarditis15,
organ involvement and fatal complications. It meningitis and renal failure16. Uveitis has
has a wide geographical distribution and occurs recently been recognized as a late complication
in tropical, subtropical and temperate climatic of leptospirosis17, 18.
zones. In the developed world most cases
that occur are associated with recreational Pulmonary haemorrhage is perhaps the most
exposure to contaminated water. The incidence fatal complication in leptospirosis. In Andamans,
seems to be increasing in developing countries. a signicantly higher case fatality ratio has
Some countries, where leptospirosis is under been observed amongst patients who develop
surveillance, have recorded this increase in pulmonary haemorrhage as compared to patients
incidence2. Most countries in the South East with other clinical presentations11. Serovar Lai
Asia region are endemic to leptospirosis. The belonging to serogroup Icterohaemorrhagiae
International Leptospirosis Society (ILS) made had been incriminated as a cause of leptospirosis
an attempt to compile data on occurrence of with haemoptysis as the predominant symptom
leptospirosis in various countries3 and the data in China and Korea19. But other serovars such as
showed that tens of thousands of severe cases Australis have also been found to be associated
occur annually world-wide. This could only be with similar clinical presentation3, 20. Serovars
an under-estimate as only a small number of Canicola and Pomona were involved in the
countries participated in the survey and even 1995 outbreak in Nicaragua21, 22. Serogroup
in those countries, leptospirosis surveillance is Canicola was responsible for an outbreak of
far from complete. leptospirosis with pulmonary haemorrhage in
Orissa, India after the super-cyclone in 19993, 8.
A number of leptospirosis outbreaks have In Andaman Islands, a few serovars belonging to
occurred during the past few years in various the serogroup Grippotyphosa including Valbuzzi
countries particularly in South America4, 5, 6 and have been isolated from cases of leptospirosis
India 7, 8, 9, 10, 11. Some of these were as a result of with pulmonary haemorrhage23.
natural calamities such as cyclone and ood.
The other most common fatal complication is
Leptospirosis is considered as the most renal failure. It was observed that a signicant
widespread zoonosis in the world12. proportion of cases with renal failure attending
Leptospirosis affects human beings and many nephrology dept. in Chennai, India had
other species of vertebrates. It can present leptospiral etiology16. However, in most cases,
in a wide spectrum of clinical manifestations renal failure is reversed with conservative
in human beings13. The syndrome of icteric measures such as maintaining uid and
leptospirosis with renal involvement is referred electrolyte balance and symptomatic therapy11.
to as Weils disease. Another recognized clinical Other complications such as meningitis rarely

Laboratory Manual 1
become fatal. Myocarditis may some times of the tropical region with plenty of rainfall
cause intractable hypotension and cardiac and it is often difcult to avoid exposure
arrhythmias and might become fatal. of the people to animals or contaminated
environment. Because of this, early case
Leptospirosis, being a zoonotic disease with detection and prompt treatment and creating
a large variety of animal species acting as awareness about the disease among the people
carriers, is difcult to eliminate and perhaps and public health professionals are the steps
even control in tropical developing countries. that could be taken to reduce the magnitude
The bacteria is adapted to the environment of the problem.

2 Leptospirosis
References
1. Waitkins SA. 1987. Leptospirosis. In: Mansons Tropical Diseases, 19th edn. Eds. Manson-Bhor PEC, Bell DR. London,
Baillire Tindall. p 657 665.
2. Tangkanakul W, Tharmaphornpil P, Plikaytis BD, Bragg S, Poonsuksombat D, Choomkasien P, Kingnate D, Ashford DA.
2000. Risk factors associated with leptospirosis in Northeastern Thailand, 1998. Am J Trop Med Hyg, 63 (3, 4): 204
208.
3. Smythe LD, Field HE, Barnett LJ, Smith CS, Dohnt MF, Symonds ML, Moore MR, Rolfe PF. 2002. Leptospiral antibodies
in ying foxes in Australia. J Wildl Dis, 38 (1): 182 6..
4. Zaki SR, Sheih WJ. 1996. Leptospirosis associated with outbreak of acute febrile illness with pulmonary haemorrhage,
Nicaragua, 1995. The epidemic working group at Ministry of Health in Nicaragua. Lancet, 347 (9000): 535 536.
5. Ko AI, Reis MG, Dourado CMR, Johnson Jr. WD, Riley LW, Salvador Leptospirosis Study Group. 1999. Urban epidemic
of severe leptospirosis in Brazil. The Lancet, 354: 820 825.
6. Barcellos C, Sabroza PC. 2001. The place behind the case: leptospirosis risk and associated environmental conditions
in a ood-related outbreak in Rio de Janeiro. Cad Sade Pblica, Rio de Janeiro, 17: 59 67.
7. World Health Organization. 2000. Leptospirosis, India - report of the investigation of a post-cyclone outbreak in
Orissa, November, 1999. Wkly Epidemiol Rec, 75: 217 223.
8. Sehgal SC, Sugunan AP, Vijayachari P. 2001. Outbreak of leptospirosis after cyclone in Orissa. National Med J India,
15 (1): 22 23.
9. Karande S, Kulkarni H, Kulkarni M, De A, Varaiya A. 2002. Leptospirosis in children in Mumbai slums. Indian J Pediatr,
69: 855 858.
10. Sehgal SC, Murhekar MV, Sugunan AP. 1995. Outbreak of leptospirosis with pulmonary involvement in North Andaman.
Indian J Med Res, 102: 9 12.
11. Singh SS, Vijayachari P, Sinha A, Sugunan AP, Rashid MA, Sehgal SC. 1999. Clinico-epidemiological study of hospitalized
cases severe leptospirosis. Indian J Med Res, 109: 94 99.
12. Faine S. 1982. Guidelines for control of leptospirosis. Geneva, World Health Organization.
13. Levett PN. 2001. Leptospirosis. Clin Microbiol Rev, 14: 296 326.
14. Silvia RRV, Brauner JS. 2002. Leptospirosis as a cause of acute respiratory failure: clinical features and outcome in
35 critical care patients. Brazilian J Infect Dis, 6 (3): 135 139.
15. Ramachandran S, Perera MVF. 1977. Cardiac and pulmonary involvement in leptospirosis. Trans Royal Soc Trop Med
Hyg, 71 (1): 56 59.
16. Muthusethupathy MA, Sivakumar S, Vijayakumar R, Jayakumar M. 1994. Renal involvement in leptospirosis our
experience in Madras city. J Post Graduate Med (India), 40 (3): 127 131.
17. Rathinamsivakumar, Ratnam S, Sureshbabu L, Natarajaseenivasan K. 1996. Leptospiral antibodies in patients with
recurrent ophthalmic involvement. Indian J Med Res, 103: 66 68.
18. Rathinam SR, Rathnam S, Selvaraj S, Dean D, Nozik RA, Namperumalsamy P. 1997. Uveitis associated with an epidemic
outbreak of leptospirosis. Am J ophthalmol, 124 (1): 71 79.
19. Oh HB, Chang WH, Cho MK, Seong WK, Park KS. 1991. Identication of new serovars yeonchon and hongchon belonging
to Leptospira interrogans of the Icterohaemorrhagiae serogroup. J. Korean Soc Microbiol 26: 253 262.
20. Simpson FG, Green KA, Haug GJ, Brookes DL. 1999. Leptospirosis associated with severe pulmonary haemorrhage in
Far North Queensland. Med J Aust 169: 151 153.
21. Zeurner RL, Bolin CA. 1997. Differentiation of Leptospira interrogans isolates by Is1500 hybridization and PCR assays.
J Clin Microbiol 35: 2612 2617.
22. Trefejo RT, Rigau-Perez JG, Ashford DA et al. 1998. Epidemic leptospirosis associated with pulmonary haemorrhage
Nicaragua, 19956. J Infect Dis 178: 1457 1463.
23. Vijayachari P, Sehgal SC, Goris MGA, Terpstra WJ, Hartskeerl RA. 2003. Leptospira interrogans serovar Valbuzzi: a
case of severe pulmonary haemorrhages in Andaman Islands. J Med Microbiol 52: 913 918.

Laboratory Manual 3
Chapter 2

Historical Perspective
S.C. SEHGAL

I
n 1886, Adolf Weil described a clinical presence of spiral organisms in kidney specimens
syndrome characterized by splenomegaly, stained with Levadeti technique (used to
jaundice, haemorrhages and nephritis1. demonstrate spirocheats) from a patient4. A
This syndrome is usually referred to as Weils World Health Organization Scientic Group on
disease and this has become synonymous with Research in Leptospirosis (1962-65) recognized
leptospirosis. Clinical syndromes resembling Stimsons description as the rst demonstration
Weils description of haemorrhagic jaundice, of leptospira5. Naguchi, who himself was not
have been known for many centuries. Such a physician, grew a spirochaete from the liver
diseases were recognized as occupational specimen of a patient, who, he was told, had
hazards of rice farmers in ancient China2. In died of yellow fever. He named this organism as
Japan, diseases with traditional names such Spirochaeta icteroids. It closely resembled the
as nanukayami (seven day fever), akiyami causative organism of Weils disease and it was
(autumn fever) or hasamiyami (autumn fever in not possible to distinguish the two organisms in
Hasami district) were later proved to be due to cross-protection tests in guinea pigs. By 1930,
leptospirosis. In Europe and Australia diseases after the discovery of yellow fever virus, it
with various local names such as cane-cutters became clear that the two organisms were
disease, swine-herds disease and Schlammeber essentially the same and the patient might have
(mud fever) were also later recognized as due had leptospirosis rather than yellow fever.
to leptospiral infection. Thus, leptospirosis had
various names in different parts of the world The Japanese workers who discovered the
that denoted seasonal association, symptoms, organism responsible for Weils disease named
duration or occupations that were thought to it Spirochaeta icterohaemorrhagiae. However,
be associated with the disease. there were considerable differences in the
appearance and movement of this organism from
Leptospires were rst identied as the cause of other spirochaetes. When Stimson observed the
Weils disease in Japan, where it was common organism in the kidney specimens, he had doubts
among coal miners3. In 1915, Inada and Ido about it being a spirochaete and designated
successfully transmitted the infection to guinea it provisionally as Spirochaeta interrogans on
pigs and from the blood of the infected animals account of its hooked extremities. The Germans
they grew the responsible organism. Unaware Huebener and Reiter called the organism
of this development, Huebener and Reiter Spirochaeta icterogenes and Uhlenhuth and
reported the successful transmission of Weils Fromme as Spirochaeta nodosa. The French,
disease to guinea pigs in October 1915. They however, adhered to the original nomenclature
demonstrated agella like bodies in Giemsa
stained blood smears. Ten days later Uhlenhuth Leptospires were rst identied
and Fromme also reported similar ndings. They as the cause of Weils disease
also recorded anicteric leptospirosis caused by
the same spirochaete for the rst time. Several
in Japan, where it was common
years before this, Stimson had reported the among coal miners.

4 Leptospirosis
Rodents are the rst leptospirosis in animals was recognized as an
important veterinary problem as well as a
recognized carriers of
source of infection to man. During the period
leptospires. A large number from 1950s to 1970s much data on the ecology
of studies on seroprevalence of leptospires in tropical countries were
generated because of military operations in
and leptospiral carrier state in
South East Asia.
rodents have been conducted
in various countries. Rodents are the rst recognized carriers of
leptospires. A large number of studies on
Spirochaeta icterohaemorrhagiae of the seroprevalence and leptospiral carrier state
Japanese authors on the ground that the in rodents have been conducted in various
Japanese workers, being the discoverers of countries. The disease in cattle was rst
the organism, were alone entitled to name identied in Russia1. A series of studies conducted
the organism. Naguchi introduced the genus by W.A. Ellis and colleagues in Northern Ireland
Leptospira in 1917 on account of the difference during 1970s and 80s revealed many aspects of
in morphology and movement. He described the transmission of the infection in animals9,
the characteristic feature of this organism 10, 11
. During the same period several studies
as long, slender, cylindrical, highly exible conducted in various countries generated a
lament with tightly set, regular, shallow lot of data on the dynamics of transmission of
spirals. The family Leptospiraceae among the the disease in various domestic animal species.
Order of Spirocheatales was proposed by Pillot It has now become clear that almost any
and Ryter in 1965. mammalian species including wild animals and
aquatic mammals can harbour leptospires and
Soon after the discovery that Weils disease was can act as source of infection to man.
caused by leptospires, several other disease
entities were recognized to have a leptospiral Initial studies on pathogenesis showed that
etiology. These include nanukayami or the leptospires are widely distributed in the organs.
Japanese seven-day fever and akiyami The studies also showed that leptospires
the harvest fever. The same Japanese group adhere to platelets and this adhesion was
that identied leprospires described the role thought to be the cause of thrombocytopaenia
of rats as their carriers3. During the early and haemorrhages seen in Weils disease.
days, it was considered that there were Information about immunity was generated
three types of leptospires i.e. Leptospira soon after the discovery of the organism and
icterohaemorrhagiae, L. icteroids and L. it was understood that immunity is mostly
hebdomadis, which differed serologically humoral in nature. In the initial publications,
from one another though morphologically they an immune agglutination with convalescent
were similar6. Leptospires were thought to be sera was described. Soon several serological
responsible for yellow fever, seven-day fever tests including latex, haemagglutination and
and Weils disease and probably for dengue other macroscopic agglutination tests were
and sand y fever6. developed. Microscopic Agglutination Test (MAT)
developed some 70 years ago still remains as
Much of the basic current knowledge about the standard. An ELISA test for the diagnosis of
leptospires and leptospirosis was understood leptospirosis was developed in 1980s12. Before
within a decade of the discovery of leptospires. the discovery of antibiotics, specic therapy was
Several types of leptospires such as limited to immunotherapy using rabbit or horse
L. icterohaemorrhagiae, L. canicola, antisera and arsenicals. Since rats were the
L. grippotyphosa, L. andamana, L. australis, rst recognized animal carrier of leptospires,
L. bataviae, L. tarassovi and L. pomona were the initial attempts at control of the disease
recognized during this period7, 8. By 1940s were centred on rodent control. Once it was

Laboratory Manual 5
understood that a wide variety of animals can reports of leptospirosis, diagnosed either by
harbour the organism and act as the source demonstration or isolation of the organisms or
of infection to man, the difculties in control by serological evidence, appeared regularly
became obvious. from several places including Calcutta, Assam
and Bombay17, 18, 19. But after the 40s, reports of
In India, reports of Weils disease had started leptospirosis were comparatively few till 1980
appearing in the literature by the end of and most the reports, which appeared during
19th century13. Chowdry reported a series of this period, were from the four metropolises.
jaundice cases that occurred over a decade
starting from 1892 in Andaman, which he Earlier, the disease was conned to a few
believed were cases of malaria, though malaria places like the Andamans, Bombay, Calcutta
parasite was not seen in blood smears. Later and Madras. During the 80s reports of human
researchers believed that these were actually leptospirosis started appearing in increasing
cases of Weils disease6, 14. Wolley reported frequency and in many places, where it
a series of 40 cases of severe malaria with had never occurred earlier. Outbreaks have
jaundice and 17 deaths that occurred among occurred in various places and often with high
self-supporting convicts of Andamans in 190915. mortality rates. Though there has not been any
In this series also, malaria parasite was absent systematic compilation of data on incidence of
in blood smear. Wolley also reported observing the disease, judged by the number of reports
motile rod like structures attached to RBCs. appearing in the literature, the disease is
In 1921 de Castro reported ve cases of toxic becoming more and more common. Now it has
jaundice or unknown origin in Andamans16. He been recognized that the disease occurs in all
examined blood lms for leptospira but failed the Southern states, in West Bengal and Assam in
to demonstrate them. In 1926, Barker reviewed the Eastern Region, in Bihar, Uttar Pradesh and
these reports and concluded that these cases Delhi in the North, in Maharashtra and Gujarat
were actually cases of Weils disease. He also in the West and in the Andaman islands. In
obtained microscopical evidence of presence of many places like the Andamans, Tamil Nadu and
leptospires in stained blood lms of patients. Kerala, leptospirosis has been recognized as an
important public health problem. Leptospirosis
In 1931, the rst report of bacteriologically accounts for considerable proportion cases with
conrmed cases of leptospirosis originated clinical complications like renal failure and
from the Andaman islands14. Twenty four myocarditis20, 21, 22. Considering the epidemic
isolates of leptospires were obtained within a potential of the disease and the nature of
period of four months from 64 patients with the environment and lifestyle of the people,
Weils disease among the free-living convicts leptospirosis is a constant threat to the health
in Port Blair and surrounding villages. Isolated of our people23.

6 Leptospirosis
References
1. Weil A. 1886. Ueber eine eigenthmliche, mit Milztumor, icterus and nephritis einhergehende, acute
infectionsskrankheit. Dtsch Arch Klin Med, 39: 209. (As quoted in Levett PN, 2001).
2. Faine S, Alder B, Bolin C, Perolat P (eds). Leptospira and Leptospirosis. 1999, Melbourne, Australia, MediSci.
3. Faine, S. 1994. Leptospira and Leptospirosis. London, CRC Press.
4. Stimson AM. 1907. Note on an organism found in yellow-fever tissue. Pub Hlth Rep, 22: 541. (As reproduced in Faine
S, 1994)
5. Abdussalam M, Alexander AD, Babudieri B, Borg-Peterson C, Eichhorn EA, Galton MM, Kaplan MM, Stableforth
AW, Turner LH, van der Hoeden J, Wolff JW, Yager RH. 1965. Classication of leptospires and recent advances in
leptospirosis. Bull Wrld Hlth Org, 32: 881 891.
6. Baker MF, Baker HJ. 1970. Pathogenic leptospires in Malaysian surface waters I. A method of survey for leptospira in
natural waters and soils. Am J Trop Med Hyg, 19: 485 492.
7. Kmety E, Dicken H. 1988. Revised list of Leptospira serovars. I Alphebetical order, II chronological order, 1 and 2,
Griningen, the Netherlands, University Press.
8. Kmety E, Dicken H. 1993. Classication of the species Leptospira interrogans and history of its serovars. Griningen,
the Netherlands, University Press.
9. Ellis WA, Logan EF, OBrien JJ, Neill SD, Ferguson HW, Hanna J. 1978. Antibodies to leptospira in the sera of aborted
bovine fetuses. Vet Rec, 103: 237 239.
10. Ellis WA, OBrien JJ, Cassells J. 1981. Role of cattle in the maintenance of Leptospira interrogans serotype hardjo
infection in Northern Ireland. Vet Rec, 108: 555 557.
11. Ellis WA, McParland PJ, Bryson DG, McNulty MS. 1985. Leptospires in pig urogenital tract and fetuses. Vet Rec,
117: 66 67.
12. Terpstra WJ, Ligthart GS, Schoone GJ. 1985. ELISA for the detection o specic IgM and IgG in human leptospirosis.
J Gen Microbiol 11: 452 457.
13. Chowdry AK. Jaundice at Port Blair, Andaman Islands. Indian Med. Gaz (1903): 38: 409 -412.
14. Taylor, J & Goyle, A.N. Leptospirosis in Andamans. Indian Medical Research Memoirs. (Memoir No. 20): Supplementary
series to the Ind. J. Med. Researdh (1931).
15. Woolley JM. Malaria in Andamans. Indian Med. Gaz (1913): 48: 266-267.
16. de Castro AB. 1922. Toxic Jaundice of unknown origin. Indian Med. Gaz. 57: 292.
17. Dasgupta, B.M. & Chopra, R.N. The occurrence of Weils disease in India. Indian Med. Gaz (1937); 72: 610-612.
18. Dasgupta, B.M. A note upon an interesting serological type of leptospira in the Andamans. Indian Med. Gaz (1939):
74: 88-89.
19. Dalal, P.M. Leptospirosis in Bombay: Report of ve cases. Indian J. Med. Sci (1960): 14: 295 301.
20. Muthusethupathy, M.A. & Sivakumar, S. Acute renal failure due to leptospirosis. J. Asso. Phy. India (1987): 35:
631-635.
21. Muthusethupathy, M.A., Sivakumar, S., Suguna Rajendar, Vijayakumar, R. & Jyaykumar, M. Leptospiral renal failure
in Madras city. Indian J. Nephro News Series (1991): 1: 15-16.
22. Ramachandran S, Perera MVF. Cardiac and pulmonary involvement in leptospirosis. Trans. Royal Soc. Trop. Med. Hyg
(1977): 71 No.1: 56-59.
23. Sehgal SC. Leptospirosis on the horizon. National medical journal of India. (2000); 13 (5): 228-30

Laboratory Manual 7
Chapter 3

Leptospira
P. VIJAYACHARI

Morphology Fig. 2. Electronmicrograph of leptospires

L
eptospires are exible helical rods that (45,0000X)
are actively motile. The motility of
leptospires is characterized by rotation
about its longitudinal axis and exion and
extension. The rotatory movement occurs in
both directions alternately. Usually one or
both ends are bent or hooked. They are too
thin to be seen under light microscope and are
best visualized under dark ground microscope.
They cannot be stained readily with aniline
dyes and can be stained only faintly by Giemsa
stain. Leptospires are best stained by silver Electron microscopic features
impregnation techniques1-3. The cell of leptospira has several coils. The
coils have amplitude of 0.1 m to 0.15 m and
Microscopic appearance under dark a wavelength of approximately 0.5 m. Freshly
ground illumination isolated pathogenic leptospires tend to be
The bacterial cells are thin, nely coiled and shorter and tightly coiled than laboratory strains
actively motile. The size of the cell ranges that have undergone repeated subcultures.
from 6-20 m in length and about 0.1 m in
thickness. Under low power magnication Leptonema illini is wider with an amplitude
(200X) bacterial cell looks like a small piece of of approximately 0.13 m and wavelength of
thread and hooked ends are visualized as dense 0.6 m whereas T. parva is shorter and tightly
dot like structures. The ne coils are seen only coiled (amplitude 0.13 m - 0.14 m and
under high power magnication (1000X). wavelength 0.3 m -0.36 m). The direction of
the coils is right handed (clock wise coiling).
Fig. 1. Leptospira under dark ground
(Microscope (200X)
Cultural characteristics
Leptospires are aerobes and utilize long chain
fatty acids as carbon and energy sources. In
addition to long chain fatty acids, Vitamin
B1, Vitamin B12 and ammonium salts are also
required for their growth. Leptospires utilize
purine bases but not pyrimidine bases and
hence they are resistant to the antibacterial
activity of the pyrimidine analogue 5- uro
uracil. This compound is used in selective
media for the isolation of leptospires from
contaminated sources.

8 Leptospirosis
Because of the inherent toxicity of free fatty i) Media that contains rabbit serum: These
acids, these must be supplied either bound include Korthofs medium, Fletchers
to albumin or in non-toxic esteried form. medium and Stuarts medium. Rabbit
Pyruvate enhances the initiation of growth serum contains nutrients including high
of the parasitic leptospires. Leptospires are concentrations of bound vitamin B12 which
grown at 28C to 30C with pH in the range of 7- helps in the growth of leptospires. All these
8. At 30C the generation time varies between media can be used for the isolation of
7-12 h and yields of 1-2 X 108 cells per ml can leptospires from the clinical specimens and
be obtained in 7-10 days. The pathogenic for the maintenance of leptospires but not
leptospires can survive for many days or even for the preparation of antigens for MAT.
months in wet soil and fresh water with neutral ii) Fatty acid albumin medium: In this medium
or slightly alkaline pH. In salt water the survival long chain fatty acid is used as a nutritional
time is only few hours. source and serum albumin as detoxicant. This
medium is popularly known as Ellinghausen-
Culture media McCullough-Johnson-Harris (EMJH) medium
A wide variety of culture media can be used for and widely used for isolation, maintenance
the cultivation of leptospires4. The routinely and preparation of antigens for MAT and for
used culture media are described briey here: growing leptospires in bulk.

Fig. 3. Phenotypic classication of leptospires

Leptospires

L. interrrogans L. biexa

Serovars (No. = 250) Serovars (No. = 45)


E.g. L. interrogans serovar E.g. L. biexa serovar
Icterohaemorrhagiae strain RGA patoc Strain Patoc 1

Serogroups (No. = 25)


Serogroups (No. = 38)
E.g. L. interrogans serovar
E.g. L. biexa serovar
Icterohaemorrhagiae strain RGA
patoc Strain Patoc 1 in
in the Icterohaemorrhagiae
the semaranga serogroup
serogroup

Laboratory Manual 9
iii) Protein-free medium: In this medium long in the presence of 8- azaguanine is being
chain fatty acids are treated with charcoal used routinely for speciation5,6. The growth
to detoxify the free fatty acids which are of pathogenic leptospires is inhibited both
highly toxic to leptospires. The antigenicity at 13C and in the presence of 8-azaguanine
and other characters of the cells grown whereas non-pathogenic ones grow at 13C
in this medium are similar to those media and are resistant to 8-azaguanine. Both the
described above. species have several serovars and serovar is
the basic taxon, which is dened on the basis
Culture media can be enriched by addition of surface antigenic makeup7,8,9. Two strains
of 1% fetal calf serum or rabbit serum to are said to belong to different serovars if
cultivate the festidious leptospiral serovars. after cross absorption with adequate amount
Selective culture media, containing 5-FU of heterologous antigen more than 10% of the
50-1000 g/ml or a combination of nalidixic acid homologous titre regularly remains in at least
50 g/ml, vancomycin 10 g/ml and polymixin B one of the two antisera in repeated tests.
sulphate 5 units/ml or a combination of actidione Closely related serovars are arranged into
100 g/ml, bacitracin 40 g/ml, 5 FU 250 g/ serogroups. However serogroup designation has
ml, neomycin sulphate 2 g/ml, polymixin B no ofcial taxonomic status and is intended for
sulphate 0.2 g/ml and refampicin 10 g/ml laboratory use.
can be used to avoid the contamination.
More than 250 serovars arranged into 25
Liquid medium can be converted into semisolid serogroups have been described under the
and solid by the addition of agar or agarose. species L.interrogans. The species L.biexa
Semisolid media contain 0.1-0.2% agar whereas has 65 serovars arranged in 38 serogroups.
solid medium contain 0.8-1% agar. Liquid The binominal classication system is strictly
medium is used for the cultivation of leptospires followed. However serovar and serogroup
to be used in various tests and other purposes. name may be added e.g. Leptospira
Semisolid medium is commonly used for the interrogans serovar icterohaemorrhagiae in
isolation of leptospires and for maintaining the serogroup icterohaemorrhagiae (L.interrogans,
cultures. Colonies are sub surface and visible at serovar Icterohaemorrhagiae in serogroup
7-21 days of incubation. Solid medium is not ideal Icterohaemorrhagiae) or Leptospira biexa
for isolation or maintenance of leptospires and serovar Patoc in serogroup Semaranga (L. biexa
mainly used for the research purpose to clone serovar Patoc in serogroup Semaranga).
the leptospires from mixed letospira cultures.
A panel of rabbit antisera (group sera) is used for
Classication of Leptospira serogroup determination. Cross Agglutination
The genera Leptospira, Leptonema and Turneria Absorption Test (CAAT) is the test of choice for
belong to the family of Leptospiraceae. The serovar determination. A panel of monoclonal
families Leptospiraceae and Spirochaetaceae antibodies (mAbs) are helpful for comparing
(genera Spriochaeta, Cristispira, Borrelia and antigenic pattern between reference strains
Treponema) make up the order Spirochaetales. and isolates.
The classication and nomenclature of
Leptospira is complex. Presently two different Genetic Classication: Based on genetic
classication systems - one based on phenotypic homology in DNA hybridization experiments, 15
characters and other on the genetic homology genomic species (L. interrogans, L. kirschneri,
are being used. L. borgpetersenii, L. santarosai, L. noguchii,
L.weilii, L. inadai, L. biexa, L. meyeri, L.
Phenotypic classication: The genus leptospira wolbachii, Genomo species 1, Genomo species
is subdivided into two species (g. 3), namely, 3, Genomo species 4 and Genomo species 5)
the L. interrogans (pathogenic) and the L. have been described in the genus Leptopsira
biexa (non-pathogenic). Growth at 13C and (g. 4) whereas Leptonema and Turneria

10 Leptospirosis
Fig. 4. Genotypic classication of leptospires

Leptospires

Species containing Species containing Species containing


pathogenic both non pathogenic non pathogenic
serovars and pathogenic serovars
serovars

L. interrogan L. inadai L. biexa


L. kirschner L. meyeri L. illini
L. borgpetersenii T. parva
L. santarosa Genomo species 5
L. noguchi
L. weili
L. wolbachi
Genomo species 1
Genomo species 3
Genomo species 4

has one species each8 (L. illini and T.parva chromosome ranges from 4,332,241 bp to
respectively). 4,277,185 bp whereas the size of the small
chromosome is in the range of 358,943 bp to
Genomic species is a group of Leptospiraceae 350,181 bp.
serovars whose DNAs show 70% or more homology
at the optimal reassociation temperature of Though the DNA-DNA hybridization is
55C or 60% or more homology at a stringent considered to be the gold standard technique
reassociation temperature of 70C and in which for species level identication of leptospires,
the related DNAs contain 5% or less unpaired it is seldom used because of its complexity.
bases. Several PCR based DNA ngerprinting methods
have become popular and are being used
The genus Leptopsira is characterized by G+C routinely for characterization of leptospires in
content of 34.4 mol%. The genus Leptonema recent years. Random Amplied Polymorphic
has G+C content of 51-53 mol% whereas the DNA (RAPD) ngerprinting, Arbitrarily Primed
G+C content of the genus Turneria is 47-48 PCR (APPCR) and Fluorescent Amplied
mol%. A complete sequencing data is available Fragment Length Polymorphism (FAFLP)
on two leptospires (serovar Lai and serovar are some of the examples. The technical
Copenhegeni). Leptospiral genome has two details of the various techniques used for the
chromosomes - large chromosome (CI) and characterization of leptospires are described
small chromosome (CII). The size of the large in chapter 8.

Laboratory Manual 11
References
1. Faine, S. 1982. Guidelines for the control of leptospirosis. WHO Offset Publication No. 67. p. 29. World Health
Organization. Geneva.
2. Faine S. (1994). Leptospira and Leptospirosis. In Taxonomy, Classication and Nomenclature. CRC Press, London. 117
140.
3. Dugui JP. Staining Methods. in Mackie & McCartney Practical Medical Microbiology, 13th ed., Vol 2 editors Collee JG,
Duguid JP, Fraser AG, Marimion BP, London: Churchill Livingstone, 1989; p. 38-63
4. Kmety E and Dikken H.. Serological typing methods of Leptospires. In Methods in Microbiology. Editors Bergan T,
Norris JR.. Academic Press, London. 1978; 11: 260-08
5. Johnson RC, Harris VG.. Differentiation of pathogenic and saprophytic leptospires:1. growth at low temperature. J
Bacteriol 1967: 94: 27 31.
6. Johnson RC, Rogers P.. Differentiation of pathogenic and saprophytic leptospires with 8-Azaguanine. J. Bacteriol.
1964; 88: 1618-23. The spirochetes in Bergeys Manual of Systematic Bacteriology., Editors : Noel R. Krieg, John G.
Holt, Williams and Wilkins, London. 1984; 1: 39-70.
7. International Committee on Systematic Bacteriology, Sub Committee on the Taxonomy Of Leptopsira, Minutes of the
meeting, 5 6 September. 1986 Manchester, England, Intl. J. Sys Bacteriol 1987; pp 472-3.
8. Brenner DJ, Kaufmann AF, Sulzer KR, Steigerwalt AG, Rogers FC., Weyant RS. Further determination of DNA relatedness
between serogroups and serovars in the family Leptospiraceae with a proposal for Leptospira alexanderi sp. nov. and
four new Leptospira genomospecies. Int J Syst Bacteriol 1999; 49 : 839-58
9. Kmety E and Dikken H. Classication of the species Leptospira interrogans and history of its serovars, 1993; University
Press Groningen, The Netherlands.

12 Leptospirosis
Chapter 4

Epidemiology of Leptospirosis
A.P. SUGUNAN

Geographic distribution and occur on the aftermath of natural calamities


magnitude of the problem such as cyclones, oods and ash-oods.

L
eptospirosis has a wide geographical
distribution and occurs in tropical, Leptospirosis is currently identied as a
subtropical and temperate climatic zones. worldwide public health problem12. In endemic
In the developed world, the incidence has come areas, leptospirosis is a major cause of various
down substantially and most cases that occur clinical syndromes such as jaundice, renal
now are associated with recreational exposure failure13, myocarditis and atypical pneumonia.
to contaminated water. In contrast, the The annual incidence of leptospirosis increased
incidence seems to be increasing in developing from 0.3/100,000 persons during the period
countries. Some countries such as Thailand, 1982 1995 to 3.3/100,000 persons in 1997
where leptospirosis is under surveillance, 98 in Thailand1. A multi-centric study in India
have recorded an increase in the incidence of showed that leptospirosis accounts for about
leptospirosis1. Most countries in the South East 12.7% of cases of acute febrile illness attending
Asia region are endemic to leptospirosis. The hospitals14. Leptospirosis is the cause of a
International Leptospirosis Society (ILS) made signicant proportion of cases of non-hepatitis
an attempt to compile data on occurrence of A E jaundice, non-malarial febrile illnesses
leptospirosis in various countries2. The data and non-dengue haemorrhagic fever in South
shows that on an average 10,000 severe cases East Asian countries15.
requiring hospitalization occur annually all over
the world. In many tropical endemic areas, a signicant
proportion of the population is exposed to
Vast majority of leptospiral infections are either leptospires. High seroprevalence rate is found
subclinical or result in very mild illness and among healthy population of many endemic
the patients may not seek medical assistance. areas such as Andaman Islands16, Seychelles
In a small proportion of patients, severe Island17, rural areas around Chennai18 etc.
complications may set in. In such cases, there High seroprevalence has also been observed
would be clinical manifestations of multiple among some of the primitive tribes living in
organ involvement and the case fatality ratio
could be more than 40 % 3. Because of the Leptospirosis is currently
protean manifestations of leptospirosis it is identied as a worldwide public
often misdiagnosed4 and under-reported.
health problem. In endemic areas,
A number of leptospirosis outbreaks have leptospirosis is a major cause of
occurred during the past few years in various various clinical syndromes such as
places such as Nicaragua5, Salvador6 and Rio de
Janeiro7 in Brazil and Orissa8, 9, Mumbai10 and
jaundice, renal failure, myocarditis
Andaman Islands11, 3 in India. Outbreaks usually and atypical pneumonia.

Laboratory Manual 13
the jungles19. Even in subtropical and inter- as carrier hosts, in whom the carrier state
tropical areas, seroprevalence could be high is temporary ranging from a few months to
among certain occupational groups such as years, and reservoir hosts in whom the carrier
farmers20,21. In endemic areas, the incidence state is life long. The reservoir hosts are the
of asymptomatic infection could also be primary source of leptospires. They infect both
very high. A survey conducted in Seychelles17 animals and human beings. The carrier hosts
showed 9% point prevalence of asymptomatic who get infection either from reservoir hosts
leptospiral infection as proved by positive or from other carrier hosts can also act as the
Polymerase Chain Reaction Test (PCR). In a source of infection to human beings. Man to
study conducted in the North Andaman, 27% man transmission is very rare and leptospiral
of the 396 persons followed up serologically infection in human beings is a dead end of
had evidence of leptospiral infection during transmission chain. Therefore, the source of
the follow up period of 12 weeks in the post- infection both in animals and humans is the
monsoon season22. carrier animal, though infection may actually
occur through various environmental vehicles.
Transmission cycle
Leptospirosis is primarily an infection of Although leptospires are susceptible to various
animals, humans being accidental victims. environmental factors, particularly drying,
The natural habitat of leptospires is the acidic pH, salinity and presence of detergents
renal tubules of carrier animals. The animal and other bactericidal agents, they can
hosts of leptospires are broadly categorized survive for long periods in water and wet soil

Fig. 1. Transmission cycle of leptospirosis

Reservoir Carrier
Hosts Hosts

Environment

14 Leptospirosis
under favourable conditions. Some serovars the environment is most suited for the survival
of leptospires have been found to survive of leptospires. In South East Asian countries,
retaining their infective potential in soil for where monsoon is active, the peak occurrence
up to 74 days23. There is also indirect evidence often follows the monsoon, when the land
that leptospires can grow and multiply in becomes wet and water-logged thus facilitating
environment under favourable conditions24. the survival of leptospires. Fig 3. shows the
Thus environment plays an important role in association of occurrence of leptospirosis with
the epidemiology of leptospirosis. rainfall in Andaman Islands. The peak occurrence
always follows the peak rainfall. In contrast,
Fig. 1 is a schematic diagram of the transmission in subtropical and temperate countries, the
cycle of leptospirosis involving reservoir hosts, peak occurrence is often observed during
carrier hosts, environment and human beings. the summer months, when the environment
becomes warm and suitable for the survival of
Three basic factors i.e. the animal carriers, the bacteria. Seasonal trends are also affected
environment suitable for the survival of by occupational or social activities such as
leptospires and exposure of people to harvesting or outdoor activities as part of fairs
contaminated environment, carrier animals, and festivals.
their urine, body uids or tissue, are always
involved in the transmission of leptospiral Carrier animals
infection. However, the exact nature of these Rats are the rst recognized carriers of
factors varies from one ecological setting to leptospires. They are often incriminated as
the other, thus making the transmission cycle the source of infection to human beings. A
complex and dynamic. Successful transmission large number of studies on seroprevalence
depends on the presence of all these three and leptospiral carrier state in rodents have
factors. Fig. 2 shows the transmission triad of been conducted in various countries. Although
leptospiral infection. Successful transmission serovar Icterohaemorrhagiae has been often
occurs in the area where all the three factors associated with rodents, other serovars have
coexist (shown in blue in the diagram). also been isolated. Other rodents such as
bandicoots25, mouse26, mongoose27, shrews28 and
Seasonal trends the aquatic rodent coypu of France29 etc. have
Occurrence of leptospirosis shows strong also been implicated as the source of infection
seasonal uctuations in most epidemiological in different ecological niches.
settings. This uctuation most often reects
seasonal changes in the environment, with peak Several domestic animal species can harbour
occurrence corresponding to the season when leptospires and act as source of infection to
human beings. Serovars Hardjobovis, Pomona,
Fig. 2. Transmission triad of leptospirosis Bratislava and Grippotyphosa are commonly
associated with cattle. Leptospirosis in cattle
could be totally unapparent or may result in
Carrier acute febrile illness, diarrhoea or mastitis.
animals Studies have shown that trans-placental
transmission of infection can occur in cattle30.
Sometimes carrier cattle could be seronegative
even to the infecting strain31 and therefore
seropositivity would not be a reliable indicator
of carrier state. In endemic areas, a signicant
Favorable Exposure proportion of the cattle could be seropositive32,
environment
which indicates that the exposure level in
cattle could be very high and they may play an
important role in the transmission of infection.

Laboratory Manual 15
Fig. 3. Association of rainfall with occurrence of leptospirosis cases in Andaman
2000 ....... Rain fall _____ Cases 80

70

1500 60

Rainfall (mm)
50

Cases
1000 40

30

500 20

10

0 0
1988 1989 1990 1991 1992 1993

Quarter

Pigs are commonly infected with serovars Transmission can be direct or indirect. Direct
Pomona, Tarassovi, Grippotyphosa, Bratislava, transmission occurs when leptospires from
Sejroe and Icterohaemorrhagiae. Adult non- tissues, body uids or urine of acutely infected
pregnant infected pigs are usually symptom or asymptomatic carrier animals enter the
free. In communities where pigs are reared body of the new host and initiate infection.
close to human habitation, they can be the Direct transmission among animals can be
source of infection to human beings. Sheep transplacental, by sexual contact or by suckling
has also been shown to harbour leptospires for milk from infected mother. Presence of leptospires
prolonged period of time33. Serovars Canicola in genital tracts as well as transplacental
and Icterohaemorrhagiae are commonly isolated transmission has been demonstrated in animals30,
from dogs. Infected dogs may suffer illness 37, 38
. Direct transmission from animals to human
usually referred as Stuttgart Disease, which beings is common in occupations that involve
is characterized by vomiting, dehydration, handling of animals or animal tissue such as
bloodstained faeces, mucosal sloughing and butchers, veterinarians, cattle and pig farmers,
death. Surviving dogs may have chronic rodent control workers etc. Accidental infection
nephritis and may shed leptospires in their to veterinarians has also been recorded39.
urine for prolonged period of time. Leptospiral
seropositivity has been observed in many wild Indirect transmission occurs when an animal
animal species including opossums34 and sea or human being acquires leptospirosis from
lions35. However, their role in the epidemiology environmental leptospires, originating in the
of human leptospirosis is not clear. urine of excretor animals. It is considered that
the most common portal of entry of leptospires
Mode of transmission and risk groups into the host body is through intact skin36. High
Leptospires are ubiquitous. They are found incidence has been recorded among people who
wherever experts in leptospirosis, medical and
veterinary practitioners and epidemiologists, Leptospiral seropositivity has been
who are aware of the epidemiology of the observed in many wild animal
disease and adequate specialist laboratory
facilities exist36. The primary source of
species including opossums and sea
leptospires is the excretor animal, from lions. However, their role in the
whose renal tubules leptospires are excreted epidemiology of human leptospirosis
into the environment with the animal urine.
is not clear.

16 Leptospirosis
are exposed to wet environments because of factors that expose people to wet environment
occupational or other activities. Leptospirosis that is likely to be contaminated with
is a known health hazard of rice farmers in leptospires as well as factors that expose
countries such as Indonesia and Thailand. High people to the excretions or tissues of carrier
incidence of leptospirosis has been recorded in animals pose risk of leptospiral infection.
provinces with large populations of farmers1. Depending on the environment and the social
Outbreaks have occurred in Korea on several and occupational behaviour, the factors that
occasions when the elds were ooded before pose risk of leptospiral infection may vary from
harvest40. Outbreaks have occurred among community to community. In a study conducted
general population when people are exposed to in North Andaman16, various outdoor activities
oodwaters that have high chance of leptospiral including agricultural work, shing in fresh
contamination8, 9, 5. water, crossing water bodies on the way and
use of stream water for bathing were found to
Leptospirosis has been recognized as a potential be associated with leptospiral seropositivity.
hazard of water sports and other recreational In Hawaii, use of water catchment systems,
activities that expose people to possibly drinking surface water and presence of skin
contaminated waters. Outbreaks associated wounds were found to be the factors associated
with recreational exposure to water have been with leptospiral infection45. In another study
reported from several countries41, 42. conducted in Northeastern Thailand, where
leptospirosis is a public health problem among
Ingestion of contaminated water has occasionally the rice farmers, various rice farming activities
been reported to cause leptospirosis by some were found to be independently associated
investigators. Cacciapouti B et al investigated with leptospiral infection1. In contrast, the risk
an outbreak of leptospirosis that occurred in factors identied in Seychelles Islands were
a small town in Central Italy. In a case-control gardening, staying in corrugated iron house, wet
analysis, drinking water from a fountain soil around house, refuse not being collected
was found to be signicantly associated with by public service, presence of cats at home,
leptospirosis43. Corwin A et al (1990), who skin wounds and drinking home brews, while
investigated an outbreak of leptospirosis at indoor occupation was found to be a protective
Okinawa, Japan, reported that swallowing factor17. Ashford DA et al studied the risk
water while swimming was signicantly factors of leptospirosis in El Sauce in Nicaragua
associated with leptospirosis44. Human-to- and came up with some interesting ndings46.
human transmission through breast-feeding The only identied risk factors were rural
has also been recorded39. However this mode household and gathering wood, while indoor
of transmission is not of much epidemiological water source was a protective factor. Shelling/
importance as such instances are rare. husking corn was found to have signicant
interaction with the type of household, with
Risk factors it being a risk factor in urban households and
Occupational, behavioural or environmental a protective factor in rural households. In a
population based matched case-control study
Occupational, behavioural or conducted in Salvador, a Northeastern coastal
environmental factors that expose city in Brazil, the identied risk factors were
related to exposure to sewage and rats47. Table
people to wet environment that
1 summarizes the risk and protective factors
is likely to be contaminated with identied in various parts of the world where
leptospires as well as factors that leptospirosis is endemic. The variability in the
risk factors in different parts stresses the point
expose people to the excretions or
that the transmission dynamics of leptospirosis
tissues of carrier animals pose risk varies greatly from one ecological setting to
of leptospiral infection. the other.

Laboratory Manual 17
Table 1. Risk factors of leptospirosis in diferent areas

PLACE RISK FACTORS PROTECTIVE FACTORS


Andaman Agricultural work, exposure to wet elds,
participating in rice harvesting, working in the
jungles, other outdoor activity, shing in fresh
water, crossing water bodies on the way, use of
stream water for bathing
Hawaii Use of water catchment systems, drinking surface
water, presence of skin wounds
Thailand Walking through water, plucking paddy sprouts for
replanting, ploughing, fertilizing in wet eld
Seychelles Gardening, staying in corrugated iron house, wet Indoor occupation
soil around house, refuse not being collected by
public service, presence of cats at home, skin
wounds, drinking home brews
Nicaragua Rural household, gathering wood Indoor water source
Brazil Open sewer in proximity to residence,
peridomiciliary sighting of rats, sighting groups
of ve or more rats, workplace exposure to
contaminants

Fig. 4. Targets for control strategies on the transmission cycle of leptospirosis

Measures to
Rodent Control make the soil Test and Slaughter
Measures and surface Test and Treat
water unsuitable Mass Treatment
for leptospires

Reservoir Host Carrier Host

Environment

Use of protective
MAN cloth at work place

18 Leptospirosis
In the rural setting it is often the carrier state is detected in animals, it could
be taken as a potential source of infection to
agricultural exposure to wet elds
humans. Several strategies have been described
possibly contaminated with the to reduce the load of carrier domestic animals.
urine of rats or farm animals which These include test and slaughter carrier
animals, test and treat, vaccination of herds
is the cause of leptospiral infection.
and rodent control measures.

Epidemiological settings Measures targeting the environment are


The eco-epidemiological settings in which application of substances to make the
leptospiral transmission occurs can be broadly environment unsuitable for the survival of
categorized as urban, rural, recreation associated leptospires, water- shed management, better
and disaster sequel. In the urban setting, rats irrigation practices etc. Measures targeting
infesting sewage networks, overowing sewage carrier animals or environment can only be
during rains, ooding of roads and exposure devised only after clearly understanding
of people to ooded roads create an ideal the transmission dynamics in a particular
environment for the transmission of leptospirosis. community, whereas measures targeting
In the rural setting it is often the agricultural people are independent of the epidemiological
exposure to wet elds possibly contaminated variability. Measures targeting human beings
with the urine of rats or farm animals which is include vaccination and chemoprophylaxis.
the cause of leptospiral infection. Leptospiral Vaccines have been developed for use on
infection also occurs due to exposure to river man48, however the existence of a large
or stream water as a result of recreational number of serovars of leptospires makes it
activities such as canoeing, rafting or swimming. difcult to develop a universally effective
Leptospirosis has now been recognized as a vaccine. Chemoprophylaxis with doxycycline
possible sequel of natural disasters such as has been tried on soldiers from non-endemic
cyclones and oods as during such times people areas visiting endemic areas and was found
and animals are exposed to wet environments to be almost 100% effective49. However, a
for a prolonged period of time. study conducted in endemic area during
an outbreak showed only 54% protection22.
Prevention and control Chemoprophylaxis can only be used in
Prevention of leptospirosis essentially is by outbreak situations or on travellers. Although
identifying the source and interrupting the the basic principles of prevention such as
transmission. Intervention strategies can reduction of source, environmental sanitation,
target many points in the transmission cycle of more hygienic work-related and personal
leptospirosis. Fig.4 summarizes the intervention practices etc. are same everywhere, there is
targets on the transmission cycle and the no universal control method applicable to all
strategies that could be adopted. In different epidemiological settings. A good understanding
epidemiological setting, different animal of the ecological, epidemiological and cultural
species could be the primary source of infection. characteristics of a community that faces
Even in the absence of epidemiological evidence the problem of leptospirosis is the essential
of association between contact with an animal prerequisite for evolving an effective and
species and human leptospirosis, if signicant acceptable intervention strategy.

Laboratory Manual 19
References
1. Tangkanakul W, Tharmaphornpil P, Plikaytis BD, Bragg S, Poonsuksombat D, Choomkasien P, Kingnate D, Ashford DA. 2000.
Risk factors associated with leptospirosis in Northeastern Thailand, 1998. Am J Trop Med Hyg, 63 (3, 4): 204 208.
2. Smythe LD. 1999. Leptospirosis Worldwide, 1999. Wkly Epidemiol Rec, 74: 237 242.
3. Singh SS, Vijayachari P, Sinha A, Sugunan AP, Rashid MA, Sehgal SC. 1999. Clinico-epidemiological study of hospitalized
cases severe leptospirosis. Indian J Med Res, 109: 94 99.
4. Flannery B, Pereira MM, Velloso LDF, Carvalho CDC, de Codes LG, Orrico, GDS, Dourado CMR, Riley LW, Reis MG, Ko AI.
2001. Referral pattern of leptospirosis cases during a large urban epidemic of dengue. Am J Trop Med Hyg, 65 (5):
657 663.
5. Zaki SR, Sheih WJ. 1996. Leptospirosis associated with outbreak of acute febrile illness with pulmonary haemorrhage,
Nicaragua, 1995. The epidemic working group at Ministry of Health in Nicaragua. The Lancet, 347 (9000): 535 536.
6. Ko AI, Reis MG, Dourado CMR, Johnson Jr. WD, Riley LW, Salvador Leptospirosis Study Group. 1999. Urban epidemic
of severe leptospirosis in Brazil. The Lancet, 354: 820 825.
7. Barcellos C, Sabroza PC. 2001. The place behind the case: leptospirosis risk and associated environmental conditions
in a ood-related outbreak in Rio de Janeiro. Cad Sade Pblica, Rio de Janeiro, 17: 59 67.
8. World Health Organization. 2000. Leptospirosis, India - report of the investigation of a post-cyclone outbreak in
Orissa, November, 1999. Wkly Epidemiol Rec, 75: 217 223.
9. Sehgal SC, Sugunan AP, Vijayachari P. 2001. Outbreak of leptospirosis after cyclone in Orissa. National Med J India,
15 (1): 22 23.
10. Karande S, Kulkarni H, Kulkarni M, De A, Varaiya A. 2002. Leptospirosis in children in Mumbai slums. Indian J Pediatr,
69: 855 858.
11. Sehgal SC, Murhekar MV, Sugunan AP. 1995. Outbreak of leptospirosis with pulmonary involvement in North Andaman.
Indian J Med Res, 102: 9 12.
12. World Health Organization. 2003. Human leptospirosis: Guidance for diagnosis, surveillance and control. Geneva,
World Health Organization.
13. Muthusethupathy MA, Sivakumar S, Suguna R, Jayakumar M, Vijayakumar R, Everard COR. 1995. Leptospirosis in
Madras a clinical and serological study. J Assoc Phy India, 43 (7): 456 458.
14. Sehgal SC, Sugunan AP, Vijayachari P. 2003. Leptospirosis: disease burden estimation and surveillance networking
in India. South East Asian J Trop Med Pub Hlth, 34, (suppl 2): 170-177
15. Laras K, Van CA, Bounlu K, Tien NTK, Olson JG, Thongchanh S. Anh TNV, Loan HK, Punjabi N, Khem HB, An US,
Insisiengmay S, Watts DM, Beecham HJ, Corwin AL. 2002. The importance of leptospirosis in Southeast Asia. Am J
Trop Med Hyg, 67 (3): 278 286.
16. Murhekar MV, Sugunan AP, Vijayachari P, Sharma S, Sehgal SC. 1998. Risk factors in the transmission of leptospiral
infection. Indian J Med Res, 107: 218 223.
17. Bovet P, Yersin C, Merien F, Davis CE, Perolat P. 1999. Factors associated with clinical leptospirosis: a population-
based case-control study in the Seychelles (Indian Ocean). Int J Epidemiol, 28: 583 590.
18. Ratnam S, Sundararaj T, Subramanian S. 1983. Serological evidence of leptospirosis in a human population following
an outbreak of the disease in cattle. Trans Royal Soc Trop Med Hyg, 77 (1): 94 98.
19. Sehgal SC, Vijayachari P, Murhekar MV, Sugunan AP, Sharma S, Singh SS. 1999. Leptospiral infection among primitive
tribes of Andaman and Nicobar Islands. Indian J Med Res, 122: 423 428.
20. Nuti M, Amaddeo D, Crovatto M, Ghionni A, Polato D, Lillini E, Pitzus E, Santini GF. 2002. Infections in Alpine environment:
Antibodies to Hantaviruses, Leptospira, Rickettsiae and Borrelia burgdorferi in dened Italian populations. Am J Trop
Med Hyg, 48 (1): 20 25.
21. Vado-Sols I, Crdenas-Marrufo ME, Jimnez_Delgaddilo B, Alzina-Lpez A, Lavida-Molina H, Suarez-Sols V, Zavala-
Velquez JE. 2002. Clinicalepidemiological study of leptospirosis in humans and reservoirs in Yuctat, Mxico. Rev
Inst Med Trop S Poulo, 44 (6): 335 340.
22. Sehgal SC, Sugunan AP, Murhekar MV, Sharma S, Vijayachari P. 2000. Randomised controlled trial of doxycycline
prophylaxis in an endemic area. Int J Antimicrob Agents, 13: 249 55.
23. Smith DJW, Self HRM. 1955. Observations on the survival of Leptospira australis A in soil and water. J Hyg, 53: 436
444. 35
24. Baker MF, Baker HJ. 1970. Pathogenic leptospires in Malaysian surface waters I. A method of survey for leptospira in
natural waters and soils. Am J Trop Med Hyg, 19: 485 492.

20 Leptospirosis
25. Saravanan R, Rajendran P, Thyagarajan SP, Smythe LD, Norris MA, Symonds ML, Dohnt MF. 2000. Leptospira autumnalis
isolated from a human case from Avadi, India, and the serovars predominance in local rat and bandicoot populations.
Ann Trop Med Parasitol, 94 (5): 503 506.
26. Collares-Pereira M, Mathias ML, Santos-Reis M, Ramalhinho MG, Duarte- Rodrigues P. 2000. Rodents and Leptospira
transmission risk in Terceira island (Azores). Eur J Epidemiol, 16 (12): 1151 1157.
27. Matthias MA, Levett PN. 2002. Leptospiral carriage by mice and mongooses on the island of Barbados. West Indian
Med J, 51 (1): 10 13.
28. Adler H, Vonstein S, Deplazes P, Stieger C, Frei R. 2002. Prevalence of Leptospira spp. in various species of small
mammals caught in an inner-city area in Switzerland. Epidemiol Infect, 128 (1): 107 109.
29. Michel V, Ruvoen-Clouet N, Menard A, Sonrier C, Fillonneau C, Rakotovao F, Ganiere JP, Andre-Fontaine G. 2001. Role
of the coypu (Myocastor coypus) in the epidemiology of leptospirosis in domestic animals and humans in France. Eur
J Epidemiol, 17 (2): 111 121.
30. Ellis WA, Logan EF, OBrien JJ, Neill SD, Ferguson HW, Hanna J. 1978. Antibodies to leptospira in the sera of aborted
bovine fetuses. Vet Rec, 103: 237 239.
31. Ellis WA, OBrien JJ, Cassells J. 1981. Role of cattle in the maintenance of Leptospira interrogans serotype hardjo
infection in Northern Ireland. Vet Rec, 108: 555 557.
32. Sharma S, Vijayachari P, Sugunan AP, Sehgal SC. 2003. Leptospital carrier rate and seroprevalence among the animal
population a cross-sectional survey in Andaman and Nicobar Islands. Epidemiol Infect, 131 (2): 985-989
33. Cousins DV, Ellis TM, Parkinson J, McGlashan CH. 1989. Evidence for sheep as a maintenance host for Leptospira
interrogans serovar hardjo. Vet Rec, 124: 123 124.
34. Ruiz-Pina HA, Puc-Franco MA, Flores-Abuxapqui J, Vado-Solis I, Cardenas- Marrufo MF. 2002. Isolation of Salmonella
enterica and serologic reactivity to Leptospira interrogans in opossums (Didelphis virginiana) from Yucatan, Mexico.
Rev Inst Med Trop Sao Paulo, 44 (4): 235 237.
35. Colagross-Schouten AM, Mazet JA, Gulland FM, Miller MA, Hietala S. 2002. Diagnosis and seroprevalence of leptospirosis
in California sea lions from coastal California. J Wildl Dis, 38 (1): 7 17.
36. Faine, S. 1994. Leptospira and Leptospirosis. London, CRC Press.
37. Ellis WA, McParland PJ, Bryson DG, McNulty MS. 1985. Leptospires in pig urogenital tract and fetuses. Vet Rec, 117:
66 67.
38. Ellis WA, Songer JG, Montgomery J, Cassells JA. 1986. Prevalence of Leptospira interrogans serovar hardjo in the
genital and urinary tracts of nonpregnant cattle. VetRrec, 118: 11 13.
39. Bolin CA, Koellner P. 1988. Human-to-human transmission of Leptospira interrogans by milk. J Infect Dis, 158: 246 247.
40. Park S, Lee, S, Rhee Y, Kang S, Kim K, Kim M, Kim K, Chang W. 1989. Leptospirosis in Chonbuk province of Korea in
1987: a study of 93 patients. Am J Trop Med Hyg, 41 (3): 345 351.
41. Anderson CD, Folland DS, Fox MD, Patton CM, Kaufmann AF. 1978. Leptospirosis: a common-source outbreak due to
leptospires of the Grippotyphosa serogroup. Am J Epidemiol, 107 (6): 538 544.
42. Sejvar J, Bancroft E, Winthrop K, Bettinger J, Bajani M, Bragg S, Shutt K, Kaiser R, Marano N, Popovic T, Tappero
J, Ashford D, Mascola L, Vugia D, Perkins B, Rosenstein N; Eco-Challenge Investigation Team. 2003. Leptospirosis in
Eco-Challenge athletes, Malaysian Borneo, 2000. Emerg Infect Dis, 9 (6): 702 707
43. Cacciapouti B, Ciceroni L, Mafeei C, Di Stanislao F, Strusi P, Calegari L, Lupidi R, Scalise G, Cagnoni G, Renga G. 1987.
A water borne outbreak of leptospirosis. Am J Epidemiol, 126 (3): 535 545.
44. Crowin A, Ryan A, Bloys W, Thomas R, Deniega B, Watts DA. 1990. A waterborne outbreak of leptospirosis among
United States military personnel in Okinawa, Japan. Int J Epidemiol, 19: 743 748.
45. Sasaki DM, Pang L, Minette HP, Wakida CK, Fujimoto WJ, Manea SJ, Kunioka R, Middleton CR. 1993. Active surveillance
and risk factors for leptospirosis in Hawaii. Am J Trop Med Hyg, 48 (1): 35 43.
46. Ashford DA, Kaiser RM, Spiegel RA, Perkins BA, Weyant RS, Bragg SL, Plikaytis B, Jarquin C, De Lose Reyes JO, Amador JJ.
2000. Asymptomatic infection and risk factors for leptospirosis in Nicaragua. Am J Trop Med Hyg, 63 (5): 249 254.
47. Sarkar U, Nascimento SF, Barbosa R, Martins R, Nuevo H, Kalafanos H, Grunstein I, Flannery G, Dias J, Riley LW,
Reis MG, Ko AI. 2002. Population-based case-control investigation of risk factors for leptospirosis during an urban
epidemic. Am J Trop Med Hyg, 66 (5): 605 610.
48. Torten M, Shenberg E, Gerichter CB, Neuman P, Klinberg MA. A new leptospiral vaccine for use in man. II. Clinical and
serological evaluation of a eld trial with volunteers. J infect Dis, 128 (5): 647 651.
49. Takafuji ET, Kirkpatrick JW, Miller RN, Karwaki JJ, Kelley PW, Gray M, McNeill KM, Timboe LH, Kane RE, Sanchez RL.
1984. An efcacy trial of doxycycline chemoprophylaxis against leptospirosis. New England J Med, 310: 497 500.

Laboratory Manual 21
Chapter 5

Clinical Manifestations
S.S. SINGH

T
he clinical manifestations of leptospirosis symptoms of restlessness, confusion, delirium
vary greatly1. The incubation period is and hallucinations and occasionally psychotic
usually 7-14 days but may range from behaviour may be prominent features in
2-21 days. Sub clinical and mild infections some patients. Some patients develop acute
are quite common. Only a small proportion of gastroenteritis with abdominal pain, vomiting
cases develop severe leptospirosis. The various and diarrhoea.
clinical manifestations are described in this
chapter. The most characteristic ndings on
examination are conjuctival suffusion and
Anicteric febrile illness severe myalgia. Conjuctival suffusion occurs
The typical leptospirosis is biphasic illness2. in the rst three days and lasts from one
The rst phase which is called as septicemic or day to more than a week. It is bilateral,
leptospiremic phase is characterized by acute most marked on the palpebral portion and is
systemic infection and by the presence of usually associated with unilateral or bilateral
leptospira in the blood and cerebrospinal uid. subconjuctival haemorrhage. There is no
This phase lasts for 4-7 days followed by a 1-3 inammatory exudate and true conjunctivitis
days of afebrile and asymptomatic period and does not occur. Myalgia is severe, even
then the second or immune phase starts with touching the muscle causes intense pain. It is
occurrence of fever and presence of leptospira most commonly observed in the lower limbs
in the urine. especially the calves. A transient macular,
maculopopular, erythematous, purpuric or
The onset is abrupt with rapidly rising fever, urticarial rash may occur, usually on the trunk
which is associated with chills and rigors, but it may be localized on the upper limbs or
increasing prostration, severe headache the shins.
and body ache. Temperature ranges from
100-105F. The headache is severe, persistent, The septicemic phase subsides after 4-7 days.
usually frontal, less often retro-orbital but The temperature then becomes normal and
occasionally may be bitemporal or occipital. the patient feels well. The second or immune
Body pains are most marked in the lower phase is characterized by severe headache
limbs especially in the calves and thighs. due to meningeal involvement and low grade
Severe pain in the back, neck, abdomen and fever. This lasts for 4-30 days or longer.
upper limbs is frequent. Prostration may be so This biphasic course may not be seen in all
marked that the patient has to stop work and patients.
frequently takes to bed. Anorexia, nausea and
vomiting are frequent and may be associated Icteric leptospirosis (Weils Disease)
with constipation or diarrhoea. Epistaxis may In some patients the septicemic phase instead
occur during the early stage. Chest pain, dry of subsiding progresses to a severe icteric
cough and haemoptysis may occur. Mental illness with renal failure. Jaundice is the most

22 Leptospirosis
important clinical feature of the severity of Anorexia and vomiting worsen
illness. Jaundice occurs between the fourth and
sixth day but may occur as early as the second
and hiccups may occur. Meningeal
day or as late as the ninth day, and deepens symptoms are frequent but
rapidly reaching a peak within a week. The liver overshadowed by hepatic and renal
is often enlarged and tender. Jaundice is due
to hepatocellular necrosis, intrahepatic
features. Confusion, restlessness,
cholestasis and increased bilirubin load from hallucinations, delusion and
absorption of tissue haemorrhage. Marked convulsions may occur.
elevations of bilirubin with mildly elevated
transaminases are characteristics. Death
rarely occurs due to hepatic failure. Renal and in severe cases the mortality may be as
involvement is the most serious complication high as 15-40%. In those who are not severely
and is the most common cause of death in ill, recovery takes place in the second week.
icteric leptospirosis. Oliguria occurs in the Diuresis occurs and the blood urea level falls
second week but may occur as early as fourth gradually. Fever subsides and the general
day of illness. A signicant number of patients conditions improve, however, jaundice takes
develop non oliguric renal failure and have a a longer time to clear.
better prognosis than oliguric renal failure.
Renal manifestations range form urinary Haemorrhagic pneumonitis
sediment changes (pyuria, albuminuria, Pulmonary haemorrhages usually occur in
hamaturia and granular casts) to severe the second week of severe forms of icteric
renal failure. Anorexia and vomiting worsen leptospirosis. Occasionally it may occur within
and hiccups may occur. Meningeal symptoms 24-48 hours of the onset of illness3. The onset
are frequent but overshadowed by hepatic is sudden with rapidly raising fever which is
and renal features. Confusion, restlessness, associated with headache, generalized body
hallucinations, delusion and convulsions may ache, and cough which is dry in the beginning
occur. Severe bleeding, cardiac and pulmonary but becomes streaked with blood after two to
complications are frequent. Towards the end three days. The patient becomes breathless
of the second week the patient is deeply and toxic. Clinical examination reveals a
jaundiced, uraemic, and haemorrhagic and toxic patient with temperature ranging
become comatose. Death may occur at this between 100-105F, tachycardia, tachypnoea
stage or early in the third week from renal and hypertension in few. Examination of the
failure. Occasionally sudden death may occur respiratory system reveals ne crepitations,
from arrythmias, cardiac failure or adrenal which in the initial stage are conned to the
haemorrhage. Massive bleeding from the bases but soon become extensive involving
alimentary and respiratory tract may also bilateral lung elds. Massive haemoptysis
end fatally. Death is rarely due to hepatic may cause asphyxiation and death. Mortality
failure but is virtually limited to icteric cases in these cases is very high and it may be as
high as 50-70% in cases who report late to the
Doxycycline has been used as hospital. Chest X-ray in these patients shows
bilateral alveolar shadowing which appears to
a chemo prophylactic agent be more dense in the mid and lower zones.
for short-term exposure, but However the radiological abnormalities may
it cannot be recommended range form a single ill dened opacity, through
multiple areas of inltration to a large area
for routine continuous use or of consolidation. These abnormalities clear
for long-term occupational up within one week without any residual
exposure. damage.

Laboratory Manual 23
Aseptic meningitis COMPLICATIONS OF LEPTOSPIROSIS
Leptospiral infection may present as meningitis
with fever, headache, photophobia and vomiting Myocarditis
and with signs of meningeal irritation (i.e. neck Cardiac complications are frequent in
stiffness and Kernigs and Brudzinskis signs). The leptospirosis. They are usually mild and are
C.S.F. Examination shows a cell count of 10 1000 observed as electrocardiographic abnormalities
cells/cumm with most of them being lymphocytes ranging from low voltage complexes, non-
and raised protein (10 200 mg/dl) with normal specic ST and T wave changes, conduction
sugar. Convulsions, focal neurological signs and defects and arrhythmias. Atrial brillations is
encephalitis are rare. Prognosis in the meningitis the most common arrhythmia. Rare but more
illness is excellent. Very rarely death may occur severe manifestations are cardiac dilatation,
form encephalitis. In Leptospiral meningitis, cardiac failure and severe arrythmias from
some distinctive features such as myalgia, haemorrhagic myocarditis. Sudden death may
conjuctival suffusion and evidence of bleeding occur from cardiac failure and arrhythmia. All
are frequently present if sought carefully. cardiac abnormalities revert back to normal
within 2-3 weeks.
Case denition
For the purpose of case detection and Haemorrhage
surveillance, a working case denition is Bleeding is a constant feature of leptospirosis
usually followed. The commonly followed case and is believed to be due to vascular damage.
denition, which is also recommended by the It is usually mild in anicteric cases but more
WHO and International Leptospirosis Society common and severe in icteric cases. Bleeding
prescribes that any person presenting with may occur from the respiratory alimentary,
acute onset of fever, headache and body aches renal and genital tracts and occasionally into
associated with: a) severe muscle tenderness, the subarachnoid space and adrenal glands.
particularly in calf muscles, b) haemorrhages Death may occur from massive bleeding.
including sub-conjunctival haemorrhage,
c) jaundice, d) cough, breathlessness and Hypotension
haemoptysis, e) oliguria or f) signs of meningeal Hypotension is an important complication noted
irritation should be suspected as a case of in patient with severe leptospirosis. The causes
leptospirosis and investigated. A suspect, of hypovolemia are: i) hypovolemia secondary
who tests positive in any of the screening to vomiting, increased insensible water
test such as dipstick, lateral ow, dri dot or losses and diminished uid intake, Ii) massive
latex agglutination test should be considered haemorrhage most often gastrointestinal,
as a probable case. Successful isolation of iii) myocardial dysfunction, iv) adrenal
leptospires from clinical specimens, a four- haemorrhage, v) widespread vascular injury
fold or higher rise in titre or seroconversion in leading to uids shifts from intravascular to
paired MAT or a positive PCR is considered as extravascular uid spaces, and vi) unidentied
conrmatory evidence of current leptospiral vocative endotoxin.
infection.
Uveitis
It is a late complication of anterior uveal tract
Bleeding is a constant feature of
and presets clinically as irritis, iridocyclitis and
leptospirosis and is believed to rarely as chorioretinitis. This may occur in the
be due to vascular damage. It is second week of illness or may be delayed up
to one year but is more frequent in the rst
usually mild in anicteric cases
6 months. Uveitis may be unilateral or bilateral
but more common and severe in and the course in variable (i.e acute benign
icteric cases. episode, recurrent episodes or a chronic process).

24 Leptospirosis
This ultimate prognosis is good but chronic uveitis and is breathless. In bacterial pneumonitis the
may cause blindness from cataract formation toxicity is not much and conjuctival suffusion
and hypopion in the anterior chamber. and muscle tenderness are absent. Pulmonary
tuberculosis and military tuberculosis have an
Leptospirosis during pregnancy insidious onset and the toxicity, breathlessness,
The hazards of leptospirosis during pregnancy conjuctival suffusion and muscle tenderness are
include intrauterine infection with foetal death not seen. In leptospirosis urine examination will
and abortion, stillbirth, premature labour and show proteinuria which will not be found in other
signs of congenital leptospirosis within a week conditions mentioned above. The chest X-ray in
or two of delivery. Leptospires may be secreted haemorrhagic pneumonitis due to leptospirosis
in the milk of lactating mothers, who during will show bilateral alveolar shadowing with clear
the septicemic phase should be regarded as apices which differentiates it from tuberculosis
potentially infective for breast fed infants. and bronchopneumonia.

DIFFERENTIAL DIAGNOSIS Aseptic meningitis has to be differentiated from


Leptospirosis with its varied manifestations may bacterial and viral meningitis and encephalitis.
mimic a large number of disease processes. Bacterial meningitis can be conrmed by CSF
examination. Viral meningitis is undistinguishable
Anicteric leptospirosis is usually misdiagnosed from Leptospiral meningitis because CSF
as P.U.O., viral fever, malaria, enteric fever, examination in both will show lymphocytes with
inuenza or pyelonephritis. The associated raised protein and normal sugar. Conjuctival
features of leptospirosis such as conjuctival suffusion, myalgia and evidence of bleeding
suffusion and muscle tenderness if carefully suggests the diagnosis of leptosirosis and
looked for may aid clinical diagnosis. Icteric this can be conrmed by serological tests. In
leptospirosis may be confused with: a) viral children clinical features that are not seen or
hepatitis, b) septicemia with jaundice, and c) rare in adults such as hypertension, a calculus
malaria. In leptospirosis the onset is abrupt, cholecystis and pancreatitis can occur.
severe headache, myalgia and conjuctival
suffusion are constant features and proteinuria TREATMENT
is common, whereas in viral hepatitis onset Antibiotic treatment is effective within 7 to
is gradual, headache and myalgia are mild 10 days of infection and it should be given
and proteinuria and conjuctival suffusion are immediately on diagnosis or suspicion. The
absent. antibiotic of choice is benzyl penicillin by
injection in doses of ve million units per day
Haemorrhagic pneumonitis can be confused with for ve days. Patients who are hypersensitive
bacterial pneumonitis, pulmonary tuberculosis to penicillin may be given erythromycin
and military tuberculosis. In haemorrhagic 250 mg four times daily for ve days.
pneumonitis the onset is quite sudden with high Doxycycline 100 mg twice daily for 10 days
fever, headache, body ache, conjuctival suffusion is also recommended. Tetracyclines are also
and muscle tenderness, patient looks quiet toxic effective but contraindicated in patients with
renal insufciency, in children and in pregnant
The antibiotic of choice is benzyl women.
penicillin by injection in doses of ve
Doxycycline has been used as a chemo
million units per day for ve days. prophylactic agent for short-term exposure4,
Patients hypersensitive to penicillin but it cannot be recommended for routine
may be given erythromycin 250 mg continuous use or for long-term occupational
exposure5.
four times daily for ve days.

Laboratory Manual 25
References
1. Faine, S. 1982. Guidelines for the control of leptospirosis. WHO Offset Publication No. 67. p. 29. World Health
Organization. Geneva.
2. Ferrar, W.E. 1990. Leptospira species (leptospirosis) p. 1813 1827. In Mandel GL, Douglas RG, Bennett JE (ed.),
Principles and Practice of Infectious Diseases. Churchill Living stone. New York
3. Singh SS, Vijayachari P, Sinha A, Sugunan AP, Rashid MA, Sehgal SC. 1999. Clinico-epidemiological study of hospitalized
cases severe leptospirosis. Indian J Med Res, 109: 94 99.
4. Takafuji ET, Kirkpatrick JW, Miller RN, Karwaki JJ, Kelley PW, Gray M, McNeill KM, Timboe LH, Kane RE, Sanchez RL.
1984. An efcacy trial of doxycycline chemoprophylaxis against leptospirosis. New England J Med, 310: 497 500.
5. Sehgal SC, Sugunan AP, Murhekar MV, Sharma S, Vijayachari P. 2000a. Randomised controlled trial of doxycycline
prophylaxis in an endemic area. Int J Antimicrob Agents, 13: 249 55.

26 Leptospirosis
Chapter 6

Laboratory Diagnosis
P. VIJAYACHARI, SAMEER SHARMA & K. NATARAJASEENIVASAN

L
eptospirosis cannot be diagnosed based a phase of leptospiraemia when the leptospires
on clinical grounds alone due to the multiply in blood and spread to different organs.
variability in clinical manifestations, The chances of recovery of leptospires from
similarity of signs and symptoms with those of blood or other tissues or body uids is usually
other bacterial, viral and parasitic infections high during this stage. This phase is followed
and frequent occurrence of the disease in by immune phase or leptospirurea phase when
atypical forms. Conrmation of the diagnosis the organisms are excreted in the urine. In
requires laboratory support. However, this phase, chances of recovery of organisms
laboratory diagnosis of leptospirosis is an area from the blood is low. The ideal specimen for
not often well understood by many of the isolation or demonstration of leptosires during
workers involved in leptospirosis diagnosis and immune phase is urine.
surveillance. Although an array of tests has been
described, availability of laboratory support, in The antibodies usually develop within 2-12 days
practice, is still a problem. Selection of the right after the onset of illness. IgM antibody starts
specimens and tests and correct interpretation appearing early in the course of the disease
of test results are important. To understand and reaches detectable levels within one week
these points rightly one should have basic or as early as on third or fourth day of illness.
knowledge regarding the sequence of events They reach peak levels during third or fourth
that occur in the host after entry of organism week and then decline slowly over months and
and antibody response against leptospires. become undetectable within six months. Rarely
IgM may persist at low level for several years.
A small number of organisms can cause
infection1. The incubation period usually ranges In a small proportion of patients (about 10%)
from 7-10 days. Leptospirosis may follow a IgM antibodies may not develop at detectable
biphasic course. During the rst 10 days, there is level and the rst appearing antibody may be

Fig. 1. Different stages of leptospirosis

Leptospiraemia

Lepto
spiru
ria

IP First Week Second Week Third Week Fourth week

Septicemic phase Immune phase

Laboratory Manual 27
Fig. 2. Level and duration of lgM antibodies at different time intervals
Titre (lgm)

0 1 2 3 4
Weeks Months Years
Duration

IgG. Hence the sera from these patients may the rst month of illness may give negative
give negative results in IgM based immuno result in Microscopic Agglutination Test (MAT).
assays. IgG antibodies appear later than IgM Serovar-specic antibodies are long-lasting than
and reach peak level after few weeks of serogroup specic antibodies. In less than 10%
illness. IgG antibody may persist at low level of patients the immune response is erratic and
for years. Microscopic agglutinating antibodies antibodies (IgM and microscopic agglutinating)
usually appear in detectable level at the end may not appear at detectable level. Hence
of the rst week of illness and reach peak negative test results observed in immuno assays
levels during third or fourth week and then do not necessarily rule out the disease among a
decline slowly over months and may become small proportion of patients.
undetectable within years or may persist at low
level even for decades. Laboratory diagnosis is broadly divided into two
categories- those that give direct evidences and
In about 10% of patients microscopic those that give indirect evidences. The direct
agglutinating antibodies appear at detectable evidences include either demonstration of
levels only after about a month of illness. Hence leptospires or its DNA and isolation of organism
the sera collected from these patients during from clinical specimens. Detection of specic

Fig. 3. Level and duration of Microscopic agglutinating antibodies at different time intervals

Genous specic antibodies


MAT titre

Serovar specic antibodies

0 1 2 3 4
Weeks Months Years
Duration

28 Leptospirosis
Fig. 4. Laboratory diagnosis Direct and indirect evidences

Laboratory diagnosis

Direct evidences Indirect evidences


Demonstration of leptospires or its DNA or Demonstration of antibodies to leptospires
isolation of organism

Microscopy Genus specic tests


Dark Ground Microscopy Macroscopic slide agglutination test
Phase contrast Microscopy Indirect haemaglution test
Counter immuno electrophoresis
Staining ELISA
Silver staining Lepto dipstick
Immunouoresence Lepto lateral ow
Lepto dridot
Isolations of leptospires
Animal Inoculation Serovar specic test
DNA Hybridization Microscopic agglutination test
Polymerase Chain Reaction

antibodies to leptospires (serological diagnosis) of this technique as a diagnostic tool has


is indirect evidence. shown that the test has low sensitivity (40.2%)
and specicity (61.5%). The usefulness of
Though several techniques have been described, differential centrifugation was limited and the
the most often used procedures and their motility of the organism further reduced after
advantages and drawbacks are mentioned here. centrifugation at higher g.

DARK GROUND MICROSCOPY Reading the results is always subjective as in the


Dark Ground Microscopy (DGM): Demonstration majority of the samples the number of organism
of leptospires by using DGM appears to be a per eld ranges from 0-2 and there was always
simple and rapid procedure. In old text books doubt about typical motility. Results obtained
it is mentioned that DGM is a useful tool for in DGM on samples from conrmed patients and
the diagnosis of leptospirosis but it is not true controls were identical. Therefore DGM is not
in practice2. Though the organism is present in recommended as a sole diagnostic tool for the
the blood during acute stage of the disease, the diagnosis of leptospirosis.
concentration is too low to allow detection by
direct microscopy. The leptospiral shedding in Specimens
urine is intermittent. Moreover serum proteins The specimens should be taken aseptically and
or cell fragments may mimic leptospires. Even sent to laboratory without delay, they must not
experienced personnel may be confused with be frozen. Oxalate, citrate or heparin may be
these artefacts as in majority of the clinical used as anticoagulant for blood or pleural uid.
samples leptospires may not exhibit typical
motility due to reactive antibodies or due Principle
to mechanical injury during the process of The principle of dark ground microscopy is that
specimen for examination. Critical evaluation the object is illuminated only by light rays that

Laboratory Manual 29
are scattered by the object. The specimen Fixative
is illuminated only by rays so oblique that Acetic acid 1 ml
unless they are scattered by the object with Formalin (40% HCHO) 2 ml
Distilled water 100 ml
a refractive index different from that of the
suspending medium the rays fail to enter the Mordant
objective and reach the eyes. Phenol 1 gm
Tannic acid 5 gm
Procedure (Blood) Distilled water 100 ml

Centrifuge 5 ml of blood (treated with an


Ammoniated silver nitrate
anticoagulant) at 1000 g for 15 min. Add 10% ammonia to 0.5% solution of silver nitrate
in distilled water until the precipitate formed just
Add approximately 10 l of plasma on a
dissolves. Now add more silver nitrate solution drop
thin microscopic slide and apply cover by drop until the precipitate returns and does not
slip. re-dissolve.

Examine under dark eld microscope with low


power and high power (X 200 and X 400).
procedures are not recommended for direct
If no leptospires are seen, centrifuge the diagnosis of leptospirosis.
plasma at 3000-4000 g for 20 min.
Carefully remove the supernatant and Fontana Staining
examine a drop of sediment microscopically
as above. Procedure
Treat the lm three times, 30 seconds each
Procedure (Urine) time, with xative.
Centrifuge a portion of freshly voided urine Wash off the xative with absolute alcohol
at 3000 g for 10 min. and allow the alcohol to act for 3 min.
Examine a drop of deposit by DGM (X 200 Drain off the excess of alcohol and carefully
and X 400). burn off the remainder until the lm is dry.
Pour on the mordant, heating till steam
Advantages
rises, and allow it to act for 30 sec.
Laboratories, where the facilities for the
other tests are not available, can undertake Wash well in distilled water and again dry
this technique. But the results should be the slide.
conrmed with other standard tests. Treat with ammoniated silver nitrate,
heating till steam rises, for 30 sec, when
Disadvantages the lm becomes brown in colour.
Low sensitivity and specicity.
Serum proteins and brin strands in blood Fig. 5. Culture stained with Fontana
resembles leptospires. Staining technique

The concentration of organism is frequently


too low in the specimens.
Requires technical expertise.
Equivocal results.

STAINING TECHNIQUES
Various silver impregnation techniques are used
for the staining of leptospires in body uids and
tissues3. However these techniques have similar
limitations as DGM. Therefore these staining

30 Leptospirosis
Wash well in distilled water, dry and mount water, to which are added 20 ml of a 2.5%
in Canada balsam. alcoholic gum mastic solution and 20 ml
It is essential that the specimen be mounted in of silver nitrate and sodium potassium
balsam under a cover slip before examination, tartarate solution). Mix the three solutions
as some immersion oils cause the lm to fade thoroughly by moving the rack with the
at once. The spirochetes are stained brownish- slides up and down for a few seconds in
black on a brownish-yellow background. the dish. Leave the slides in the reduction
mixture for 12-15 min. until the colour of
the sections changes to light brown.
Stock Solutions Wash 3 times in distilled water, dehydrate,
Silver nitrate solution
clear in xylol, and mount in neutral mounting
Silver nitrate 10 gms. in 1000 ml distilled water.
Place in an amber coloured bottle with glass uid.
stopper and store in refrigerator.

Alcoholic gum mastic


The spirochetes appear with a black mirror
Gum mastic 2.5 gms. in 100 ml absolute ethanol. like contrast distinguishable from the yellow
Dissolve by stirring and leave to stand overnight, or brown tissue elements.
lter several times until clear.

Silver nitrate and sodium potassium tartrate


ISOLATION OF LEPTOSPIRES
Silver nitrate 2 gms., sodium potassium tartarate Isolation of leptospires from clinical specimens
1.65 gms in 1000 ml distilled water. Dissolve the is the strongest evidence for conrmatory
silver nitrate in boiling distilled water. Quickly add diagnosis. Isolation and identication is the
the sodium potassium tartarate until the white
method of choice to identify circulating
precipitate changes to gray. Filter while hot into an
amber coloured bottle and store in refrigerator. serovars in a particular geographical region. In
addition locally isolated and identied strains
Hydroquinone solution will be more useful to be used as antigens in
Hydroquinone 1 gm in 60 ml double distilled
MAT as local strains were found to be more
water.
sensitive and strongly reactive than reference
strains. Moreover the local strains will be used
MODIFIED STEINER AND STEINER for the development of vaccine. However the
TECHNIQUE technique has several drawbacks. Leptospires
are slow growing organisms and require several
Procedure days or weeks to yield cultures and weeks to
Six to seven m thick sections from formalin months for identication. Prior administration
xed tissue blocks embedded in parafn of antibiotics greatly reduces the chances of
should be cut and mounted on slides with successful isolation.
egg albumin.
De-parafnize and hydrate in the usual
Blood Culture
Ideal time : Within 10 days of the onset of
manner.
symptoms.
Immerse in a 1.0% aqueous solution of silver Media : EMJH and Fletchers media.
nitrate for 2 hours at 56-58C in an oven.
Wash thoroughly in distilled water. Procedure
Swab the area with spirit.
Dehydrate up to absolute alcohol and place
the slides in 2.5% of alcoholic gum mastic Draw blood using sterile syringe and needle
solution for 5 min. by vein puncture.
Transfer from the gum mastic solution into Inoculate two and four drops into two tubes
a reducing solution of hydroquinone (1 gm containing 5 ml medium.
hydroquinone dissolved in 60 ml distilled Incubate at 30C for 4-6 weeks.

Laboratory Manual 31
Examine the culture using dark eld The above procedure should be repeated two
illumination initially on rst, third and fth or three times with urine samples collected
days followed by 7-10 days interval up to 6 on different days to increase the probability
weeks. of isolation.

Selective culture media containing 50-1000 g/ Urine can be ltered (through 0.22 m lter)
ml of 5-FU or a combination of nalidixic acid 50 and/or inoculated into selective culture media
g/ml, vancomycin 10 g/ml and polymixin B to avoid contamination.
sulphate 5 units/ml or a combination of actidione
100 g/ml, bacitracin 40 g/ml, 5-FU 250 g/ Urine may have acidic pH in many cases.
ml, neomycin sulphate 2 g/ml, polymixin B Therefore urine should be collected in tubes
sulphate 0.2 g/ml and refampicin 10 g/ml containing equal amount of PBS with pH 7.2.
can be used to avoid the contamination.
CSF CULTURE
Subculture should be made within 48 hours to Time : 5-10 days of the onset of the
minimize the inhibitory effect of the selective disease.
agents on leptospires. Medium : As above

Growth of the fastidious isolate is encouraged by Procedure


adding to the medium 0.1% to 0.15% agarose and Inoculate 0.5 ml of CSF into 5 ml of culture
0.4% to 1% of rabbit serum or fetal calf serum. media.
Follow the same procedure as blood culture.
URINE CULTURE
Time : 10-30 days after the onset of the Advantages of isolating leptospires from
disease. clinical specimens
Media : Same as above. Denite proof of infection.

Circulating serovars can be identied.


Procedure
Collect a sample of midstream urine (freshly Local isolates can be used as antigens in
voided midstream urine within 2 hours is MAT.
the ideal specimen). Local isolates can be used in vaccine
Dilute the urine as follows using sterile development.
test tubes and sterile phosphate buffer pH
7.2.pH 7.2. Disadvantages
Fastidious organism requires special
Add 0.4 ml of urine to 3.6 ml of PBS pH 7.2
medium for isolation.
(1 in 10).
Leptospires grow slowly. Isolation of
Add 3 ml of (a) to 3 ml of PBS (1 in 20).
leptospires from clinical specimens takes
Add 2 ml of (b) to 2 ml of PBS (1 in 40). several days to several weeks.
Add 1 ml of (c) to 1 ml of PBS (1 in 80). The technique is laborious, time consuming
Each resulting 0.5 ml dilution of above is and is not possible in small laboratories.
inoculated into 4 separate 5 ml volume of Contamination of culture media by
medium. other micro-organisms or by saprophytic
Label the tubes mentioning the dilution. leptospires is common in routine practice.
Incubate at 30C. The successful isolation rate is less due to
prior use of antibiotics, imperfectly cleaned
Examine the culture using dark eld
glass ware or wrong incubation temperature
illumination at an interval of 7-10 days up
and pH.
to six weeks.

32 Leptospirosis
ANIMAL INOCULATION tubes are incubated at 30C. Cultures are
Clinical materials may be inoculated into examined by Dark Field Microscope (DFM).
laboratory animals. Leptospires in the clinical If they are satisfactory the cultures are
materials may cause disease in the animals, dispensed into an Erlenmeyer ask containing
if they are susceptible. Leptospires can be 50 ml of EMJH medium and then incubated
observed directly in the peritoneal uid or for 5-7 days at 30C in a shaking incubator.
cultured from the peritoneal uid, blood or Formalin is added to nal concentration of
tissues. 0.5% and the cultures are allowed to stand
for at least 30 min.
Animals used
The cultures are centrifuged for 30 min at
Hamsters (4-6 weeks old)
10,000 g.
Guinea Pigs (150-175 gms.)
The supernatant is discarded and the tubes
Procedure are allowed to dry in a slanted position for
The inoculation of blood and/or urine is two hours.
done intra-peritoneally (0.5-1.0 ml) The sediment is re-suspended in 1.5 to 2 ml
From the third day after inoculation, of solution containing 0.5% formalin, 12%
peritoneal uid is examined under Dark Field NaCl and 20% glycerol in distilled water and
Microscope for the presence of leptospires. mixed 1520 times by sucking up and down
with a 2 ml syringe through a 1820 gauge
The examination should be done during needle.
3-10 days after inoculation.
The antigen is kept at 4C overnight.
When the inoculated animals die, it is
possible to isolate leptospires from liver Measure the optical density at 520
and kidneys. nanometers in a spectrophotometer. Adjust
to a range of 0.550-0.600 by diluting
Animal inoculation is not recommended due to suspension with the NaCl - Formalin-
ethical reasons. Maintenance of animals is very Glycerol solution.
expensive. Standardize the antigen using control sera.
Store at 4C.
MACROSCOPIC SLIDE
AGGLUTINATION TEST (MSAT) Procedure
The basic principle of the test is similar to other
Place 10 l serum on a glass slide. Add one
slide agglutination tests used in other infectious
drop (50 l) of antigen and mix thoroughly
diseases such as enteric fever or brucellosis.
with a applicator stick.
There can be different ways to prepare the Place the slide on the mechanical rotator
antigens. One may either use a single serovar and rotate at 120 rpm for 4 min and read
or multiple serovars to prepare the antigens. the reaction with an indirect light against a
When multiple serovars are used as antigens, black background.
antigens can be pooled. Since the technique
will require lot of laboratory work, generally Recording the results
a single non-pathogenic serovar patoc (strain The reaction is recorded as ++ when the clumps
Patoc 1) is used. are large and denite, + when the smaller
clumps are well dened but the suspension
Preparation of the Antigen is not clear, +/- when ne clumps are visible
Leptospires are grown in liquid medium but the suspension is not clear and negative
preferably EMJH. Tubes with 3 ml medium when the mixture in the drop is unchanged.
are inoculated with the few drops of a 5-7 Agglutination of + and ++ is considered as
days old leptospira culture. The inoculated positive.

Laboratory Manual 33
The test is easy to perform and read. The antigen consumed. The turbidity disappears quickly
is broadly reactive and stable for six months at upon cooling. If there is turbidity, leave the
4C to 8C. It is more sensitive than Microscopic cultures a few days longer in the shaking
Agglutination Test (MAT) in the early stage of incubator until all of the Tween has been
the disease. However a high percentage of false used up.
positive reactions are observed, probably due After ensuring the quality of the culture
to lack of standardization and quality control of and absence of Tween kill the leptospires
the antigen preparation. The number of false with formalin (0.5% nal conc.).
negative reactions are comparatively low.
After one hour heat the killed leptospira
ENZYME IMMUNO ASSAY (EIA) culture in boiling water for 30 min, shaking
EIA is one of the techniques commonly used every 5 min.
for the diagnosis. The test can detect specic After cooling to room temperature,
antibodies earlier than MAT. The advantage of centrifuge the culture for 30 min. at 10,000
the test is that it can differentiate between g. Centrifugation at lesser g is also possible.
recent and past infection by detecting the The supernatant is used as antigen.
type of antibodies (IgM or IgG) present in the In the liquid form, the antigen is stable for
clinical specimen. In this test, broadly, reactive years if stored at 4C.
antigen is used. The antigen antibody reaction is
visualised or measured by spectrophotometer/ Coating of the ELISA Plates
ELISA reader using a conjugate (enzyme 100 l of antigen is pipetted into every well
conjugated to anti-IgM or IgG) and a colour of the ELISA plate. A sample of each batch
reagent. The main drawback of the test is it of plates should be checked before being
should be standardized locally to nd out cut used for routine diagnosis.
off titre or signicant titre for current infection
The plates are left at room temperature
as once a man is infected with leptospires, IgM
until complete evaporation of the uid
may persist at low level for several years.
has taken place (1-3 days, depending on
Preparation of the antigen for temperature). Avoid exposure to sunlight.
the EIA The coated plates are kept in a closed box, or
Leptospira interrogans serovar Copenhageni in sealed plastic bags, at room temperature.
strain Wijnberg or L. biexa serovar Patoc The antigen is stable for several years.
strain Patoc I is grown in EMJH medium for
10-12 days at 30C in shaking incubator. Procedure
Abundant growth is necessary to produce a Rinse the coated plates four times with
good antigen. PBS/Tween.
If necessary, add 5 ml of sterile diluted During the last two washings leave the uid
Tween 80 (1:10) to 500 ml culture under in the wells for one minute.
sterile conditions 4-5 days after inoculation. Dry the plates by tapping them on a cloth or
Incubate for another 5-6 days. lter paper.
Before harvesting, check for abundant Make doubling dilutions of the patients and
growth and purity. Also check for the controls sera in PBS/Tween/BSA. Fill the
absence of Tween. This can be done by second column of the wells with 190 l of
adding formalin to a sample from the dilution uid, the other wells with 100 l.
culture to get a nal concentration of Add 10 l of patients and controls serum
0.5%, centrifuging at 10,000 g for 30 min to the second column (dilution is now 1:20).
and heating the supernatant in a glass tube Transfer, after mixing thoroughly, 100 l to
in a boiling water bath. If the supernatant the 3rd well, etc. Discard the nal 100 l.
becomes turbid, not all the Tween has been Use the rst column as blank.

34 Leptospirosis
Allow rapid processing of large number of
PBS pH 7.2
samples.
Dissolve per litre distilled water
8.5 g NaCl
0.85 g Na2HPO4 2H20 Disadvantages
0.25 g KH2PO4 Infecting serovar cannot be assessed.
PBS/Tween Calibration of cut off value and signicant
PBS pH 7.2 + 0.05% Tween 20
PBS/Tween/BSA
titre are required.
PBS/Tween + 0.5% BSA Comparatively less specic.
Preparation of the Reagents:
Conjugate
Equipment required
Dilute the conjugate in PBS/Tween/BSA. For
Micropipettes (Multi & Uni Channel)
each conjugate an optimal dilution should be
Microtips
used (i.e. block titration)
Pipettes
Substrate
Dissolve 80 mg 5-amino-2-salicylic acid in 100
ml distilled water at 75C. This solution is stable Comments: A titre of 1:40 in non-endemic
for 1 month if kept at 40C. Before use bring pH areas and a titre of 1:80 in endemic areas is
to 6.0 with 1 N NaOH. Add 9 parts of 5-AS 1 part
of 0.05% H2O2 just before use. (1.5 ml 1% H2O2 to
considered as signicant titre for leptospirosis
28.5 ml distilled water) in early stage.

MICROCAPSULE AGGLUTINATION
Cover the plate with another microtitre TEST (MCAT)
plate and incubate for one hour at 30C. The test is based on the principle of passive
After one hour wash the plate as under 1. agglutination and employs carrier micro-capsule
Add to all wells 100 l conjugate, diluted in particles on the surface of which ultrasonicated
PBS/Tween/BSA. For each of the conjugates leptospiral antigens are absorbed.
an optimal dilution should be used (i.e.
block titration).
Kit
The kit contains two vials of lyophilized reagents
Incubate for one hour at 30C as before. viz reagent A and reagent B and one vial of
Discard the contents and wash four times as diluents, a test tube rack with mirror for reading
above. the results, test tubes (small) and disposable
Add to all wells 100 l substrate. loop (1 l). Reagent A incorporates antigens of
L. australis, L. autumnalis and L. hebdomadis
Leave at RT for two hours.
and reagent B incorporates that of L. canicola,
L. icterohaemorrhagiae and L. pyrogenes.
Reading the results
Read the optical densities using a multi
Procedure
scanner with a 492 nm lter. Use column
Add 2.3 ml of reconstituting solution to the
one for measuring blank.
vials of A and B.
The end point (titre) is the last well (dilution)
Place 0.3 ml of reagent A into one test tube
in which you nd a colour giving an OD equal
and 0.3 ml of B reagent into another.
to or greater than half of the value of the
darkest well of the positive control. Using a loop for 1 l take about 1 l, of the
test serum and place it into the test tube
Advantages containing reagent A and B separately.
Detects IgM antibodies earlier than MAT. Shake the test tubes and allow to stand
Single antigenic preparation can be used. them in the test tube rack with mirror.

Heat stable antigens which are stable at After three hours read the results.
room temperature for long periods.

Laboratory Manual 35
Reading the results detection reagent in the test tube and
The results are recorded as follows - positive mix.
when the agglutination pattern cover the Wet the white test strip of the dipstick by
entire bottom of the tube, doubtful when immersing the white end of the dipstick
agglutination pattern is slightly larger than the briey in the dipstick uid (vial C).
negative control and negative if no agglutination
Incubate the white end of the dipstick
is observed.
in the mixture of detection reagent and
Advantages serum. Remove any air-bubbles by moving
Simple to perform and easy to read. the dipstick up and down. Incubate for
three hours at room temperature.
Does not require any special expertise or
equipment. Remove the dipstick from the detection
reagent and rinse with water. Allow to dry.
Disadvantages
Not a conrmatory test. Kit
Reconstitution uid (vial A)
Kits have to be imported.
Lyophilized detection reagent (vial B)
Costlier. Dipstick uid (vial C)
Dipsticks containing, on a white strip, a leptospira
antigen (lower band) and an internal control band
Fig. 6. Positive and negative results in MCAT (upper band)

Equipment
Test tubes
Test tube holder
Micropipettes (5-250 l)
Disposable pipette tips

Interpretation of the results


a) Positive: Development of clearly visible
reddish coloured Ag band (1+, 2+, 3+, 4+).
b) Negative: No development of reddish
coloured Ag band.

LEPTO LATERAL FLOW


Lepto lateral ow is based on the binding of
LEPTO DIPSTICK
specic IgM antibodies to the broadly reactive
Principle heat extracted antigen prepared from non-
The assay is based on the binding of leptospira- pathogenic Patoc 1 strain. IgM antibodies bound
specic IgM antibodies to the leptospira to the broadly reactive antigen are detected
antigen. Bound IgM antibodies are detected with an anti human IgM gold conjugate
with an anti-human dye conjugate5. contained within the test device6.

Procedure Kit

Reconstitute the lyophilized reagent (vial Lepto lateral ow device


Sample uid (diluent)
B) with 5 ml reconstitution uid (vial A).
Transfer 200 l reconstituted detection Equipment
reagent to a marked test tube. Micropipettes (10 l)
Disposable pipette tips
Add 4 l serum to the reconstituted

36 Leptospirosis
Procedure suspended by mixing with the serum sample
Add ve ml of undiluted serum or 10 using a special spatula followed by swirling of the
l of whole blood added to the sample suspension by hand rotating of the agglutination
application. card. Serum, which has been stored frozen, can
Add 130 l of sample uid (diluent). be used as well. Agglutination occurs within
30 to 60 seconds and is clearly visible by the
Wait for up to 15 min. formation of ne aggregates that tend to settle
at the edge of the droplet7.
Reading results
If only the control band became stained,
Kit
the test is negative. Lepto dri dot cards
If both test and control bands became Disposable plastic spatulas
stained, the test is considered as positive.
Equipment
Micropipettes (10 l)
Advantages Disposable pipette tips
Very quick.
Both serum as well as blood can be used to Procedure
perform the test. Remove a Dri-Dot card from the packaging
and place the card on a bench top with the
Disadvantages blue dot facing upwards.
Expensive. Spot 10 l serum next to, but not onto, the
blue dot and within the area marked by the
Fig. 7. Lepto lateral ow, positive and black circle.
negative test results Take hold of the plastic spatula with the
at site of the tip facing downwards. Hold
the spatula with the thumb and forenger
close to the at end of the spatula. Suspend
serum and blue dot with a quick circular
motion while pressing the at end of the
spatula rmly on to the dot. Dont spread
the suspension outside the area marked by
the black circle. Proceed with the next step
when the blue dot is fully suspended and a
homogenous suspension is obtained.
LEPTO DRI-DOT Keeping the card near horizontally, slowly
rotate the card swirling the liquid in circular
Principle motion within the limits of the marked areas
The Lepto Dri-Dot consists of coloured latex in order to mix latex and serum sample
particles activated with a broadly reactive further and to induce agglutination.
leptospira antigen that is dried onto an Read results within 60 seconds.
agglutination card. The assay is based on the
binding of leptospira-specic antibodies to Advantages
the leptospira antigen. The broadly reactive Simple to perform and easy to read.
antigen ensures the efcient detection of a
Doesnt require any special expertise or
wide spectrum of leptospira infections. The
equipment.
assay is performed by the addition of 10 l of
a freshly prepared serum sample to the dried The Dri-Dot have long shelf lives even at
latex particles. The latex particles are then room temperature.

Laboratory Manual 37
Disadvantages Rotate the slide slowly and gently.
Although the results of the Lepto Dri-Dot Record the results within 60 seconds.
are found to be in agreement with those of
IgM ELISA, neither 100% sensitivity or 100%
Fig. 8. Lepto LAT, positive and negative
specicity is claimed. test results
It is advised that the MicroscopicAgglutination
Test to be used as a conrmatory test.
Kits have to be imported.
Costlier.

LEPTO-LAT
LeptoLAT is a latex agglutination test for
detection of antibodies to leptospires in body
uids.
Interpretation of the test results
Fine clearly visible agglutination occurs
Materials provided
within 30-60 seconds. The intensity of the
1. Sensitized latex beads in suspension
2. A positive control agglutination depends on concentration of the
3. A negative control antibodies in a serum sample. Clearly visible
4. Plastic sticks for mixing granular agglutination is indicates the presence
of specic antibodies to leptospires. In stronger
Materials required but not provided
1. Clean glass slides (Please do not use already used reactions ne granular clumps tend to settle at
ones) the edge of the circle. Agglutination that occurs
2. Micro pipette (5 l 10 l) beyond 60 seconds may be due to evaporation
3. Micropipette tips
and should not be considered.

Storage
Principle
The coated latex particles are stable up to one
Lepto LAT is a agglutination immuno assay for the
year at 4C 8C but can be stored for up to six
detection of leptopsira specic antibodies. The
months at room temperature (3237C).
blue coloured latex beads are sensitized with the
broadly reactive and specic antigen prepared
Advantages
from pathogenic leptospires and suspended in
Simple to perform and easy to read and
storage buffer. When the specimen is mixed
doesnt require any special equipment or
with suspended latex, antibodies present in the
expertise
specimen interact with the antigen that is coated
on the surface of the latex particles leading to The Lepto-LAT have long shelf life even at
the formation of ne and clearly visible granular room temperature
agglutination within 60 seconds. Cost effective.

Procedure Disadvantages
Shake well the vial containing coloured Results need to be conrmed.
latex beads.
Transfer 10 ul of latex beads on a slide.
MICROSCOPIC AGGLUTINATION
TEST (MAT)
Add 5 l of serum near to the latex. The most commonly used serological technique
Mix the serum thoroughly with the latex for conrmation of diagnosis is MAT. The test
beads and spread uniformly in circular is highly sensitive when performed on paired
fashion. (The diameter of the circle should sera (acute and convalescent ) and is serovar/
not be more than 1 cm). serogroup specic. One of the critical issues

38 Leptospirosis
of MAT is the cut-off or signicant titre for The optimal cut-off titre is assessed by carrying
diagnosis, when the test is done only on a single out a baseline study on distribution of titres
sample8. Ideally MAT should be performed in the community as well as among conrmed
on paired sera collected during acute and patients. Using this data it is possible to estimate
convalescent stage of the disease to nd the sensitivities and false positivity rates at
out sero-conversion or four-fold rise in titre, different cut-off titres. The titre that gives the
which is the evidence of current or recent lowest number of false results is then chosen as
infection. However, collection of convalescent the optimal cut-off titre. This titre can be found
serum sample is difcult in routine practice. out by plotting false positivity rates against
It also delays the diagnosis. The alternative sensitivities at different titres. Such a plot is
is to determine cut-off or signicant titre called the Receiver Operating Characteristics
for MAT on a single sample for diagnosis. The (ROC) curve. The titre that lies closest to the
cut-off titre for single MAT depends on the left upper corner of the plot will result in least
baseline titre in the community in a particular number of false results i.e. sum of false positive
geographical region. Different laboratories results and false negative results and hence is
use different cut off titres ranging from 1 in 100 considered as the optimal one. Figures 11 and 12
to 1 in 400 for diagnosis based on endemicity show ROC curves for MAT during rst week and
or baseline titre in the community. Several second to fourth weeks of illness in endemic and
investigators usually consider a titre of 1 in non-endemic areas. In an ROC curve, a straight
100 as a signicant titre for diagnosis without line drawn from left lower corner to right upper
considering the endemicity or baseline titres corner is called the no-benet line, because if a
in the community. This may result in over- titre that lies below this line is chosen as cut-off
diagnosis and overestimation of disease the test result doesnt provide any information
burden. useful for diagnosis.

Fig. 9. ROC curve of MAT cut-off titres in endemic area showing that a titre of 1 in 200 is the
optimal during 2-4 weeks and no titre is optimal during rst week

100%
1:100 1:50
1:200

80%
1:400

60%
Sensitivity

1:800

1:50

40%
First week
1:1600 1:100
2-4 weeks
No benet
20%
1:3200
1:200
1:400
1:800
0%
0.0% 20.0% 40.0% 60.0% 80.0% 100.0%

False Positivity Rate (1-specicity)

Laboratory Manual 39
Fig. 10. ROC curve of MAT cut-off titres in non-endemic area showing that a titre of 1 in 100 is
the optimal during 2-4 weeks and 1 in 50 during rst week

100%

1:50
1:100

80% 1:200

1:800

60%
Sensitivity

1:50

40%
1:100 First week
2-4 weeks
No benet
1:20
20%

1:400

0%
0.0% 20.0% 40.0% 60.0% 80.0% 100.0%

False Positivity Rate (1-specicity)

Although MAT has several drawbacks, it is free non-agglutinable leptospires. Hence, there
an indispensable technique in diagnosis and is an element of subjectivity in reading the test
in seroepidemiological studies. The test is results.
more specic and sensitive if performed on
paired sera. However, this is always not true Panel of antigens
and therefore, one should be careful while A battery of antigens, covering the range
interpreting MAT titres. Due to cross reactive of serovars that are expected or likely to
antigens among serovars belonging to different be circulating in a particular geographical
serogroups or due to the lack of homogenicity area, where the patient becomes infected,
within the serovars of certain serogroups, MAT should be used. Locally isolated strains
titres may not always provide actual information should be included in the panel, if possible,
about infecting/circulating serogroups. as they may give more specic and sensitive
results than reference strains. At least
The usual method for carrying out MAT is to one strain of saprophytic serovar (Patoc I)
mix equal volumes of series of serum dilutions should also be included in the panel to act
and leptospira culture in the wells of microtitre as genus specic antigen to detect infections
plates. The serum antigen mixture is allowed caused by serovars/strains not yet known to
to react for a certain period at certain exist in a particular geographical area. The
temperature. The degree of agglutination and recommended panel of antigens (one strain
endpoint titre are determined by examining a representing each known serogroup) may
drop of the mixture by dark eld microscopy. The be used while testing sera received from a
degree of agglutination as such may be difcult geographical area where the information
to observe and the degree of agglutination is about circulating serovars is lacking. The
often assessed by determining the number of recommended panel is given below.

40 Leptospirosis
Preparation of antigens Formalinized antigens
The stock culture collection of leptospires is The live cultures killed by addition of a nal
best maintained in screw-capped test tubes 0.5% formaldehyde solution may be used as
containing 5-6 ml of liquid medium. Fresh sub antigens. These antigens are stable for at least
culture are made by inoculating 0.5 ml from two months. The disadvantage of these antigens
the each strain/serovar into tubes. At the is that leptospires tend to adhere loosely to
same time, a loop-full of the culture should be one another, thus interfering with the reading
examined by dark eld microscopy to conrm of the reaction.
the presence of viable leptospires and the
absence of contamination and it should be Density of the antigens
free from breeding nests. The inoculated Well grown culture, a minimum density of
cultures are incubated at 30C and checked for 1-2 x 108 leptospires per ml should be used as
the presence of the growth after 5-7 days. The an antigen. The density can be determined by
cultures used as antigens should be checked by direct counting, using spectrophotometer or by
MAT against homologous antisera frequently for McFarlands scale.
quality control.
Procedure
Live antigens Fill all 96 wells of microtitre plate with
The culture may be used directly as the antigen. 50 l PBS.
The antigens can be used up to one week after Add another 140 l PBS to the wells of
5-7 days of incubation. column 2.

Table 1. Commonly used serovars/strains for MAT

SEROGROUP SEROVAR STRAIN


Andamana Andamana CH 11
Australis Australis Ballico
Bratislava Jez Bratislava
Autumnalis Autumnalis Akiyami
Rachmati Rachmat
Ballum Ballum S 102
Bataviae Bataviae V Tienen
Canicola Canicola Hond Utrecht IV
Celledoni Celledoni Celledoni
Cynopteri Cynopteri 3522 C
Grippotyposa Grippotyposa Moskva V
Hebdomadis Hebdomadis Hebdomadis
Icterohaemorrhagiae Icterohaemorrhagiae RGA
Copenhageni M 20
Javanica Javanica Veldrat Bat. 46
Poi Poi
Panama Panama CZ 214
Pomona Pomona Pomona
Pyrogenes Pyrogenes Salinem
Shermani Shermani LT 821
Sejroe Sejroe M 84
hardjo Hardjoprajitno
Semaranga Patoc Patoc 1
Tarassovi Tarassovi Perepelitsin

Laboratory Manual 41
Add 10 l of serum to the wells of column Advantages
2 (dilution is now 1:20) mix and discard It is serovar/serogroup specic test. Some
100 l. clue about the infecting serovar can be
Dilute by pipetting 50 l from one well to obtained.
the next, discard the nal 50 l (2- >3- >4- It has sensitivity and excellent specicity .
>5- >6- >7- >8- >9- >10- >11- >12). Once infected, the person stays MAT Positive
Add 50 l leptospira culture to all wells. for several years. So the test is useful for
Mix thoroughly on a micro shaker. epidemiological purpose.

Incubate for 2-4 hours at 30C. Disadvantages


1421 strains have to be maintained in
Reading of the test results culture, which is often very difcult.
The serum-antigen mixtures are examined
under a dark eld microscope for agglutination. Procedure is complex and time consuming.
For observation, one drop of mixture is Reading results requires experienced
transferred with a platinum loop or pipette personnel.
from a well to a microscopic slide and examined It is not possible to distinguish between IgM
under dark eld microscope with 20X objective antibodies indicative of current infection,
without cover slip. and IgG antibodies indicative of past
infection.
The titre is the dilution that gives 50%
agglutination, leaving 50% of cells free. Finding of agglutinating antibodies in a
Compare with a control suspension of single serum sample does not necessarily
leptospires diluted one in two in PBS without prove current leptospirosis. An antibody
serum (column 1) titre may be due to residual antibodies
of a past infection. Therefore, the
Notice that with killed antigen observed interpretation of a single titre is not easy
agglutination is measured directly, contrary so a second serum sample is required for
to the procedure to the live antigen where, demonstrating a raising titre which has a
essentially, agglutination is measured diagnostic signicance.
indirectly by establishing the reduction of Calibration of signicant titre is essential in
leptospiral density with 50% in comparison the case of single serum specimens.
with the density of free leptospires in the
False negativity in the early course of the
control.
disease.
Note: Serum samples will be stored at 20C.
Comments: In the early stage of the disease
Prior to examination, serum samples will
cross reactivity may occur and patient sera
be checked for the presence of debris or
may agglutinate a variety of other serovars.
contamination by bacteria that may interfere
Sometimes even to higher titres than the
with agglutination.
causative serovar, a phenomenon is called
as paradoxical reaction. However, such
Criteria for serological diagnosis of
paradoxical reactions tend to drop in titre and
leptospirosis by using MAT
usually the homologous titre to the causative
Seroconversion or four fold rise in antibody
organisms predominate in later stages of the
titre in paired sera.
disease.
A minimum titre of 1:400 or more in a single
Other serological methods include Immuno
serum sample. However, the signicant titre in
Fluorescent Antibody test (IFA) and Indirect
the case of single serum samples may vary from
Haemagglutination test (IHA). Different
one geographical area to other.

42 Leptospirosis
Fig. 12. Microscopic Agglutination Test (MAT) End titre 1 in 160

Negative control 1 in 20

1 in 40
1 in 80

1 in 160 1 in 320

methods used in the laboratory for the diagnosis Four-fold or greater rise in the MAT titre
of leptospirosis can also be categorized as between acute and convalescent-phase
microscopic, bacteriological, immunological/ serum specimens run in parallel.
serological or molecular biological (g. 14). Sero-conversion from a titre < 1 in 20 to 1
in 80 in between acute and convalescent-
Criteria for a denite diagnosis of phase samples run in parallel.
Leptospirosis
The criterion for laboratory diagnosis varies Isolation of leptospires is laborious and takes
from one laboratory to the other. The criteria several weeks or months. Sero-conversion or
usually used in well established laboratories rise in titre is the central dogma of serological
are mentioned below: diagnosis but it requires second convalescent-
Isolation of leptospires from a clinical phase sample which is difcult to obtain.
specimen. Therefore, the criteria for denite diagnosis has

Laboratory Manual 43
Fig. 13. Laboratory diagnosis different approaches

Laboratory diagnosis

Bacteriologic Microscopic Immunologic Molecular

Isolation Direct Microscopy Microscopic Polymerase


agglutination test chain
Animal Immuno
reaction
Inoculation histochemical ELISA
(PCR)
straining
Indirect
In situ
Immunoorescence haemagglutination
hybridization
test
Silver Inpregaation
techniques Lepto dipstick

Lepto lateral ow

Lepto dri-dot

greater application in establishing the endemicity assays, slide agglutination test or latex
of the disease in a particular geographical agglutination test.
region rather than in routine diagnosis. Once Demonstration of leptospires directly or by
the endemicity is dened in a particular region, staining methods.
criteria for a presumptive diagnosis can be used
for diagnosis and case management. No single MAT titre can be regarded as diagnostic
of acute or active infection and sometimes
Criteria for a Presumptive conrmed patients have low MAT titres. Cross
Diagnosis reactive antibodies in syphilis, relapsing fever,
A MAT titre of 100/200/400 or above in lyme disease, enteric fever, dengue and malaria
single sample based on endemicity. may give a titre of 80 or 100. Therefore low MAT
A positive result in IgM based immuno- titres need to be interpreted with caution.

44 Leptospirosis
References
1. Faine, S. 1982. Guidelines for the control of leptospirosis. WHO Offset Publication No. 67. p. 29. World Health
Organization. Geneva.
2. Vijayachari, P., Sugunan, A.P, Umapati, T and Sehgal, S.C. 2001: Evaluation of Dark ground microscopy as a rapid
diagnostic procedure in leptospirosis. Indian J Med Res. 114: 54-58
3. Dugui JP. Staining Methods. in Mackie & McCartney Practical Medical Microbiology, 13th ed., Vol 2 editors Collee JG,
Duguid JP, Fraser AG, Marimion BP, London: Churchill Livingstone, 1989; p. 38-63
4. Sehgal, S.C., 1997: Vijayachari, P. and Subramaniam, V. Evaluation of Leptospira Micro Capsular Agglutination Test
(MCAT) for sero-diagnosis of Leptospirosis. Indian J. Med. Res. 106:504
5. Sehgal, S.C., Vijayachari, P., Sharma, S., Sugunan, A. P. 1999: LeptoDipstick - a rapid and simple method for
serodiagnosis of leptospirosis in acute stage. Trans. Roy. Soc. Trop. Med. Hyg. 93:161-164
6. Sehgal, S.C., Vijayachari, P., Sugunan, A.P., Umapathi, T. 2003: Field application of Lepto lateral-ow for rapid
diagnosis of leptospirosis. Journal of Medical Microbiology, 52, 1-6
7. Vijayachari, P, Sugunan, A.P and Sehgal, S.C. 2002: Evaluation of Lepto dri-dot as a rapid test for the diagnosis of
leptospirosis. Epidemiology & Infection 129, 617-21
8. Vijayachari. P, Sugunan, A.P, Sehgal, S.C. 2001: Role of Microscopic Agglutination Test (MAT) as a diagnostic tool
during acute stage of leptospirosis in low and high endemic areas. Indian J. Med. Res. 114: 99-106

Laboratory Manual 45
Chapter 7

Serological Characterization
of Leptospires
P. VIJAYACHARI, SAMEER SHARMA & K. NATARAJASEENIVASAN

T
he taxonomic status of leptospira can be 8-Azaguanine Test
dened by a combination of its various
characteristics1. Therefore different Procedure
markers should be considered or tests should Make suspension of 100 ml distilled water
be repeated. The scheme used for classication and 225 mg. 8-Azaguanine
and identication of leptospires is shown in Sterilize the ask with the 8-Azaguanine
Table 1. solution at 121C for 30 min.
Add aseptically 0.5 ml 8-Azaguanine solution
Table 1: Scheme used for classication and to 4.5 ml EMJH media
identication of leptospires Mix thoroughly
Inoculate tubes with 0.05 ml of a well-grown
Taxonomic Main Characteristic or Test used
culture of the strain under investigation and
Status
the control
1. Genus morphology, movement
Incubate at 30C
2. Species growth at 13C and growth with 8- Check growth twice in a week up to 21
azaguanine (225 g/ml), days
3. Serogroup agglutination with group sera

4. Serovar a) agglutination test and agglutinin Controls


absorption tests Pathogenic strains with and without 8-
b) factor analysis with factor sera.
Azaguanine
5. Genomic a) DNA Hybridization Saprophytic strains with and without 8-
Species b) RFLP Azaguanine
c) RAPD
d) REA
13C Test
Differentiation of pathogenic and Procedure
saprophytic leptospires Inoculate 0.05 ml of a well grown culture
On the basis of morphological characteristics, into 5 ml of EMJH medium of the strain
pathogenic leptospires cannot be under investigation and the controls in
differentiated from saprophytic ones. The duplicate.
commonly used tests to discriminate between Incubate one set at 13C and the other at
saprophytic and pathogenic leptospires grow 30C
at low temperature (13C) and resistant Check twice in a week by dark eld
to 8-Azaguanine (225 g/ml). Pathogenic microscopy for growth
leptospires does not grow at 13C and they Used controls (Pathogenic strains and
are resistant to 8-Azaguanine. Saprophytic strains)

46 Leptospirosis
Serogroup Serovar Strain Titre
1. Australis australis Ballico Negative
2. Australis bratislave Jez Brat. Negative
3. Autumnalis bangkinang Bangkinang I 1:80
4. Autumnalis butembo Butembo Negative
5. Autumnalis carlos 3C Negative
6. Autumnalis rachmati Rachmat Negative
7. Ballum ballum Mus 127 Negative
8. Ballum kenya Njenga Negative
9. Bataviae bataviae Swart Negative
10. Canicola canicola H.Uterecht IV Negative
11. Canicola schueffneri VI.90 C Negative
12. Celledoni cellodoni Cellodoni Negative
13. Cynopteri cynopteri 3522 C Negative
14. Djasiman djasiman Djasinman 1:40
15. Grippotyphosa grippotyphosa Moskva V 1:10240
16. Grippotyphosa huanuco M4 1:160
17. Hebdomadis hebdomadis Hebdomadis Negative
18. Hebdomadis worsfoldi Worsfold Negative
19. Icterohaem. copenhageni M 20 Negative
20. Icterohaem. icterohaem RGA Negative
21. Javanica poi Poi Negative
22. Louisiana louisiana LSU 1945 Negative
23. Manhao manhao L 60 Negative
24. Mini mini Sari Negative
25. Panama panama CZ 214 K Negative
26. Pomona pomona Pomona Negative
27. Pyrogenes pyrogenes Salinem Negative
28. Sarmin rio Rr 5 Negative
29. Sarmin weaveri CZ 390 Negative
30. Sejroe hardjo Hardjopraj Negative
31. Sejroe saxkoebing Mus 24 Negative
32. Shermani shermani 1342 K Negative
33. Tarassovi bakeri LT 79 Negative
34. Tarassovi mogden Compton Negative
35. Tarassovi rama 316 Negative
36. Tarassovi tarassovi Perepelicin Negative
Strain DS2 probably belongs to serogroup Grippotyphosa

DETERMINATION OF SEROGROUP of a certain serogroup but in general gives little


Presently there are more than 230 pathogenic cross reactions with strains of other serogroups.
serovars, placed into 25 serogroups. To Table 2 shows the results obtained in MAT with
determine group reactivity, the unknown strain X group sera on an isolate D-15.
has to be tested in the Microscopic Agglutination
Test (MAT) with rabbit antisera group sera. A MAT with positive groups
representative group serum is a selected rabbit If the unknown strain (X) reacts positively
antiserum, which reacts strongly with all serovars with one or more group sera the MAT will be

Laboratory Manual 47
performed with all reference antisera which the serovar. The methods used for this purpose
belong to the positive group(s). If necessary an include the Cross Agglutination Absorption Test
antiserum against the unknown strain (X) can (CAAT), the factor sera analysis, typing based
be produced in a rabbit. The antiserum has to on Monoclonal antibodies (serological) and
be tested in the MAT with all reference strains Restriction Enzyme nuclease Analysis (REA),
which belong to the positive group. Restriction Fragment Length Polymorphism
(RFLP) technique, RandomAmplied Polymorphic
Calculation of the results DNA (RAPD) ngerprinting technique.
Cross agglutination tires are expressed as
percentages of the reciprocal titre for the Cross Agglutination and Absorption
homologous strains of positive antisera from Test (CAAT)
the whole serogroup with the unknown strain Two strains are considered to belong to different
(X) and vice versa. Two strains are considered serotypes/serovars if after cross-absorption
to be related in some way if the agglutination with adequate amounts of heterologous antigen
titre of unknown strain (X) with an antiserum 10% or more of the homologous titre regularly
and/or the agglutination titre of antiserum (X) remains in at least one of the two antisera in
with reference strain are more than 10%. Titres repeated tests.
are expressed using the formula
The amount of antigen to be used for absorption
Tgs is a very important factor (Wolff and Broom,
Tui = x 100 1954; Kmety et al 1970). It is important that
Ths the quantitative relationship between antigen
and immune serum to be absorbed should be
where Tui is the agglutination titre of the well balanced to avoid the possible non-specic
unknown strain, Tgs is the reciprocal titre of absorption by excessive amounts of antigen
antisera from positive group(s) with unknown (babudieri, 1971). Therefore variable amounts
strain and Ths is the reciprocal of reference of antigen are mixed with the antiserum. This
antiserum with homologous strain and antiserum is standardized to a MAT titre of
1:5120. The absorbed antisera whose titre
Tgi with the absorbing strain is approaching zero
Tus = x 100 (should not exceed 1% of the pre-absorption
Thi titre) is used for the agglutination with the
homologous strain. Tests to be repeated
where Tus is the agglutination titre of unknown several times.
sera, Tgi is the reciprocal titre of unknown sera
with reference strains from positive groups and All cross agglutination titres of more than 10%
Thi is the reciprocal titre of the unknown serum are considered to be positive
with homologous strain.
Perform absorption test with
Strain DS-15 and bananal are unrelated as in All relevant reference antisera that react
both sera less than 10% homology is observed. positively with the unknown strain X.
Strain DS-15, canalzona and grippotyphosa are All relevant reference strains that react
related as in both sera more than 10% homology positively with the produced antiserum
was observed (Table 3). against unknown strain (X).

DETERMINATION OF SEROVAR The following control test have to


STATUS be performed prior to absorption
The serovar is the basic taxon in the taxonomy of test
leptospires2,3,4. So it is important to characterize In the cross-absorption test, those
an unknown strain of leptospira to the level of pre-dilutions of antisera are used, which give

48 Leptospirosis
a MAT titre of 1:5120 with the homologous 20 ml culture) in 1 ml PBS-formalin diluted
killed antigen. antiserum each.
If the MAT titre of the antiserum is Absorption of the antiserum with the
higher than 1:5120, dilute to the desired sediments has to be performed overnight at
concentration. 30C.
Add 0.4 ml pre-diluted antiserum to 3.6 ml Spin down the leptospires by centrifugation
PBS pH 7.2 with formalin (0.5%). for 30 minutes at 10,000 g.
3 ml of this diluted antiserum is used for The supernatants are pipetted off
the absorption tests with different amounts carefully.
of sedimented leptospires and 1 ml is used The supernatants are used for the MAT.
for the homologous control test.
Agglutination after absorption
Inoculation and centrifugation To check for over or under absorption, the
Inoculate a well-grown culture into 50 ml absorbed reference antisera are tested
EMJH medium. in the MAT with live and killed antigen
Incubate in the shaking incubator for 5-7 (absorbing strain X).
days at 30C. Fill all the wells of microtitre plate with 50
Check the culture by dark eld l of PBS pH 7.2.
examination for the absence of debris and
Add 50 l each of the absorbed
contamination.
antiserum to be tested to the wells of
Treat the well-grown cultures (minimum
column 2. The dilution in this well is now
density = 2 x 108 leptospires/ml) with
1:20.
formalin (0.5% nal concentration).
The formalinized cultures are allowed to Add to all wells 50 l antigen
stand for one hour. (absorbing stain X). After the addition of
Divide the culture over 5, 10 and 20 ml the antigen the dilution will be 1:40 in
leptospira quantities and centrifuge the column 2.
three different amounts of leptospira After reading the MAT one has to chose
culture for 30 min. at 10,000 g. on from the three absorbed antiserum
After centrifugation the supernatants have for further testing. Take the absorbed
to be discarded. antiserum each having a MAT titre of 1:40
or 1:80. This titre has to be less than
Absorption 1% of the homologous titre for the same
Re-suspend the sediments with air-dried antiserum.
cells of strain X (derived from 5, 10 and

Table 3: Cross agglutination absorption test results on isolate DS-15

Antiserum Abs. Homologous Titre Relation


Strain before absorption after absorption
Canalzonae DS-15 L 5120 L 5120 100
K 5120 K 5120 100

DS-15 Canalzonae L 10240 L 5120 50


K 5120 K 2560 50
Grippotyphosa DS-15 L 10240 L 320 3.1
K 5120 K 160 3.1

DS-15 Grippotyphosa L 2560 L 40 6.2


K 2560 K 80 3.1

Laboratory Manual 49
With the chosen absorbed Calculation of the results
antiserum the following MATs are
performed Interpretation of results
Control test of the unabsorbed diluted Relationship between strain DS-15 and
reference antiserum with live and killed canalzona: In both sera more than 10% of the
homologous reference antigen. homologous titre remains. They belong to the
different serovars.
Test of the absorbed serum with live and
killed homologous reference antigen.
Relationship between strain DS-15 and
The tests can also be performed with the grippotyphosa: Both strains are related as after
produced antiserum against strain X with absorption both the serum samples give less
all positive reference strains. than 10% homologous titre.

Equipment TYPING USING MONOCLONAL


Disposable microtitre plates (96 wells) ANTIBODIES (mAbs)
Pipettes Mouse monoclonal antibodies (mAbs) are used
Plastic tips
to type isolates as belong to certain serovars5.
Incubator 30C
Multichannel diluter
Characterization can be done by the use of
Dark eld microscope (objectives 16-20 X, eye MCAs that react with a single characteristic
pieces 10 X) epitope or with several MCAs that react with a
Microscopic slides characteristic mosaic of epitopes.

Reagents Histograms
Phosphate Buffered Saline (PBS), pH 7.2
Leptospira reference strains and Isolates Serovars can be identied by their characteristic
Mouse monoclonal antisera antigenic pattern recognized by a set of
Leptospires of control reference strains and isolates monoclonal antibodies which are called
will be grown in EMJH medium, incubated for 5-7
histograms.
days at 30C and checked by dark eld microscopy
for adequate density and absence of debris or
contaminating bacteria.

50 Leptospirosis
References
1. Faine, S. 1982. Guidelines for the control of leptospirosis. WHO Offset Publication No. 67. p. 29. World Health
Organization. Geneva.
2. Kmety E and Dikken H.. Serological typing methods of Leptospires. In Methods in Microbiology. Editors Bergan T,
Norris JR.. Academic Press, London. 1978; 11: 260-08
3. International Committee on Systematic Bacteriology, Sub Committee on the Taxonomy Of Leptopsira, Minutes of the
meeting, 5 6 September. 1986 Manchester, England, Intl. J. Sys Bacteriol 1987; pp 472-3.
4. Kmety E and Dikken H. Classication of the species Leptospira interrogans and history of its serovars, 1993; University
Press Groningen, The Netherlands.
5. Korver H, Kolk AHJ, Vingerhoed J, Van Leeuwen J, Terpstra WJ. Classication of serovars of the Icterohaemorrhagiae
serogroup by monoclonal antibodies. Isr J Vet Med 1988; 44: 15-18.

Laboratory Manual 51
Chapter 8
Molecular Tools in the
Diagnosis and Characterization
of Leptospires
D. BISWAS & SUBARNA ROY

R
ecently, DNA-based techniques have popularity, the use of labeled probes for
been proposed as alternative methods molecular diagnostics has diminished.
of diagnosis and identication of
leptospires. Nucleic acid probes and Polymerase Chain Reaction (PCR)
hybridization techniques were used previously PCR method involves in vitro amplication
for detection of leptospires from clinical of genus-specic target DNA sequence, if
samples1,2. Polymerase Chain Reaction (PCR) present, in clinical samples. A pair of short
was subsequently developed for detection of DNA fragments, known as primers is used for
leptospires from clinical samples. Different specic amplication of DNA fragments from
groups developed different primers targeting the pathogen in blood, urine or CSF. Positive
different locations of the leptospiral genome diagnosis results from the amplication of the
for sequence-specic amplication of target sequence whereas negative samples
sequences in PCR that holds promise as a fail to produce amplied DNA in PCR. PCR
molecular tool for diagnosis of the disease3, can be used to detect leptospiral infection in
4, 5
. PCR is very often used as a very sensitive both animals and human beings following the
and specic test for diagnosis of many diseases same methods. Van Eys et al7 developed PCR
and PCR with different primers specic for for detection of leptospires in urine samples
Leptospira is in the process of evaluation in of infected cattle. Urine samples containing
different laboratories across the world. as few as 10 leptospires per ml gave positive
results in PCR assay. Gravekamp et al4 proposed
Hybridization with Nucleic acid the use of two sets of primers (G1 & G2 and
probes B641 & B651) that enabled the amplication of
Nucleic acid hybridization can conrm target DNA fragment from leptospiral species.
diagnosis before the results of culture and Amplication results in generation of a PCR
biochemical tests are available. Dot blot product of 285 bp and could detect even 1-10
hybridization method with 32P- and biotin- leptospires per ml of urine. Leptospires were
labeled genotype-specic probes has been also detected from aqueous humor of a patient
used for early detection of leptospires6. In with unilateral uveitis using silica particles
situ hybridization method using biotin-labelled and guanidine thiocyanate method by PCR9. It
DNA probe for detection of L.interrogans in has been found that PCR is a rapid, sensitive
clinical samples has also been proposed. In a and specic means of diagnosing leptospiral
particular study, it was found that nucleic acid infection, especially during the rst few days
hybridization detected 60 of 75 urine samples, of the disease when antibodies are not fully
whereas FAT detected 24 samples and only detectable in serological tests. In different
13 samples were detected by bacteriological studies involving patients suffering clinically
culture. However, hybridization with nucleic with acute leptospirosis, PCR has been compared
acid probes is tedious and costly and with with bacterial culture and MAT for diagnosis
polymerase chain reaction gaining more and it has been concluded that the PCR is an

52 Leptospirosis
efcient tool for early diagnosis of leptospirosis
during acute phase of the disease, especially Materials and Equipment
1. Tris
when the clinical symptoms are confusing. 2. Guanididne thiocyanate
The most important advantages of PCR are 3. EDTA
its sensitivity, specicity and rapidity through 4. Triton x 100
which the disease can be diagnosed. However, 5. Sodium hydroxide
6. Kieselguhr-DG
the major drawbacks of this technique are 7. Proteinase K
its high operational cost and unavailability of 8. Ethidium bromide
facilities in common diagnostic laboratories. 9. Taq DNA Polymerase
10. Assay buffer for Taq DNA Polymerase
11. dNTPs mixture
The detection of leptospires in the clinical
12. Sets of primers (specic for Leptospira)
samples by using PCR is one of the most 13. Mineral oil
valuable additions in the laboratory for
diagnosis of leptospirosis. PCR is an iterative
process consisting of three elements: Equipment
1. PCR thermo cycler
2. Submarine horizontal gel electrophoresis
1. Denaturation of the template DNA into two apparatus
separate strands at an optimal temperature 3. UV transilluminator
and time depending on their G+C content 4. Vorter mixer
5. Water bath shaker
and size of the template DNA.
6. Micro centrifuge
2. Annealing of the oligonucleotide primers to 7. Eppendorf micro centrifuge tube
the single stranded sequences at an optimal 8. PCR tube
9. Micropipettes and mcrotips
temperature of 3-5C lower than the 10. Camera and lm
calculated melting temperature at which
the oligonucleotide primers dissociate from
their templates. thoroughly at room temperature for
3. Extension of the annealed primers with 10 min.
the help of dNTPs and catalyzed by the Centrifuge the mixture for 2 min. at 16,000
thermostable DNA polymerase. However, g and sediment the diatoms.
number of cycles required for amplication Wash the diatoms with absorbed DNA twice
of DNA depends on the number of copies with 1 ml of L2 wash buffer [120 g of GuSCN
of template DNA present at the time of in 100 ml of 0.1 M Tris Hcl (pH 6.4)], twice
beginning of the reaction and the efciency with 1 ml of 70% (V/V) ethanol and once
of primer extension and amplication. Once with 1 ml of acetone.
the reaction starts it proceeds until one of
Dry the diatoms pellets at 56C.
the components becomes limiting.
Elute the DNA in 120 l of double distilled
Methods H2O for 10 min at 56C in presence of 5 l
of 10 mg/ml Proteinase K.
Serum or Plasma preparation for PCR
Inactivate the Proteinase K by incubating
Mix serum sample with L6 lysis buffer [120
the sample at 100C for 15 min.
g of GuSCN, 22 ml of 0.2 M EDTA, 2.6 ml of
Triton X 100 in 100 ml of 0.1 M Tris Hcl (pH Centrifuge for 2 min at 16,000 g.
6.4) in the ratio of 1:9 (V/V)]. Use 10 to 40 l of eluted DNA sample for PCR.
Add 40 l of diatom suspension [0.5 ml of 36%
(W/V) HCl and 10 g of diatoms (Kieselguhr-
Urine Sample Preparation for PCR
Collect 10 ml of Urine in a 15 ml container
DG) in 50 ml of H2O].
and add 10 l formalin (nal concentration
Incubate the mixture after vortexing 0.1%) and store the sample at 4C till use.

Laboratory Manual 53
Centrifuge 10 ml of Urine for 30 min at Fig. 1. Lines 1-5, 8-11:285 bp fragment
1500 g. amplied by G1 & G2 (all genospecies except
L. Krichneri); Lanes 7-8:563 bp fragment
Discard the supernatant.
amplied by B641 & II (L. Krischneri)
Add 9 ml of L6 lysis buffer and 40 l of
diatoms or silica to the pellet.
Incubate by shaking for 10 min at RT.
Centrifuge for 5 min at 1500 g.
Discard supernatant by suction.
Add 1 ml of L2 wash buffer and transfer the
suspension to a clean 1.5 ml tube.
Wash the pellets twice with 1 ml L2 wash
buffer (1 min at 16000 g).
Wash the pellets twice with 1 ml 70% ethanol
(1 min at 16000 g).
Wash the pellets once with 1 ml acetone
(1 min 16000 g).
Dry the pallets for 10 min at 56C.
Elute the DNA in 120 l of double distilled
H2O for 10 min. at 56C in presence of 5 l
of 10 mg/ml Proteinase K.
Boil the eluted DNA solution for 10 min at cycler for 30 cycles. Each cycle consists
100C. of denaturation of DNA at 94C for
Centrifuge for two min at 16,000 g. 1 min, annealing of the primer at 55C for
1 - 2 min and extension for two min. at
Collect 100 l supernatant with a pipette
72C. After the cycles are completed, the
and use 10 to 40 l for the PCR reaction.
nal extension of the amplied product is
done at 72C for seven min.
Amplication of DNA
The amplication of DNA is performed in a The amplied PCR products are analysed
total volume of 50 l. in 1% - 2% agarose gel with 0.1 g/ml
ethidium bromide in TAE buffer pH 8.0.
The primers used for the PCR are G1 5-
At the end of the electrophoresis the gel
CTG AAT CGC TGT ATA AAA GT-3 & G2 5-
is visualized under UV transilluminator
GGA AAA CAA ATG GTC GGA AG-3 and B64I
and photographed. Amplication of 285
5- CTG AAT TCT CAT CTC AAC TC-3 & B64II
/563base pair DNA fragment indicates
5 GCA GAA ATC AGA TGG ACG AT-3
presence of leptospiral DNA in the
The reaction mixture (50 l ) contains specimen.
5 l of 10x assay buffer [10 mM Tris Hcl
(pH 9.0), 1.5 mM MgCl2, 50 mM Kcl and Advantages
0.01% Gelatin], 200 M each dNTPs, Gives relatively quick results in the early
20 pM of each primer, 0.5 U of Taq DNA stage of the disease when antibodies have
Polymerase, Template DNA (10-40 l). not yet developed in detectable levels.
Add 20 l of mineral oil on top of the
reaction mixture, if required, to prevent Disadvantages
evaporation. Complicated and expensive
Amplication is carried out in a thermal Sophisticated equipment are required.

54 Leptospirosis
MOLECULAR CHARACTERIZATION Amplied Fragment Length Polymorphism
Conventionally leptospires are classied (AFLP).
serologically on antigenic properties with Pulsed Field Gel Electrophoresis (PFGE).
a serovar as the basic taxon. However, the
DNA sequence analysis.
Microscopic Agglutination Test (MAT), Cross
Agglutination Absorption Test (CAAT) and
monoclonal antibody (mAbs) techniques that
ARBITRARILY PRIMED PCR (AP-PCR)/
are used for serological characterizations are RANDOM AMPLIFIED POLYMORPHIC
generally laborious, time-consuming, and often DNA (RAPD) FINGERPRINTING
cannot distinguish all serovars. Moreover, tests
like the MAT and CAAT require ready supply of Materials and equipment required

live antigens. Tris-Hcl pH .8, 10 mM Magnesium chloride, 4 mM


Taq DNA polymerase KCl 50 mM
With the advent of modern molecular biological Thermal cycler Tris EDTA buffer
tools, several attempts have recently been Electrophoresis apparatus Ethidium bromide
made to characterize leptospires by these UV Transilluminator Agarose type II
techniques. Most of these techniques are Vortex mixture RAPD primers
DNA-based. DNA-based characterization Water bath shaker dNTPs
scheme is based on the genetic make up and can Micropipettes Gel documentation system
translate the genetic code into visible patterns. Microtips
These methods are highly reproducible and can
show true genetic afnities and relationships Principle
between isolates. The AP-PCR, also called random amplied
polymorphic DNA ngerprinting technique is
Some of the methods that are currently being based on the amplication of the DNA in the
used in characterization of leptospires are: PCR by short random oligonucleotide primers.
Arbitrarily primed PCR (AP- PCR) or The size and number of the amplied products
Randomly Amplied Polymorphic DNA depend on the particular primers and template
(RAPD) ngerprinting. DNA. It is used to compare intra and inter-
Restriction Endonuclease Analysis (REA). specic differences among pathogens including
Restriction Fragment Length Polymorphism leptospires. The RAPD technique can be applied
(RFLP). on puried DNA, crude extracts of cells from
cultures or from colonies on agar plates. This
Fig. 2. RAPD ngerprints of reference of technique is being used for the molecular
Lines 1-5, 8-11:285 bp fragment amplied differentiation/typing of various leptospiral
by G1& G2 (all genospecies except L. isolates/strains.
Krischneri); Lanes 7-8:563 bp fragment
amplied by 8641 & II (L. Krischneri) Procedure
DNA for AP-PCR is prepared following the
method of Boom et al9. AP-PCR ngerprinting
technique is carried out as per the method
described by Roy et al10. M16, a 22-mer
oligonucleotide (5 AAA GAA GGA CTC AGC
GAC TGC G 3) previously used as one of a pair
of primers for amplication of an insertion
sequence in Leptospira genome, is used as a
primer for AP-PCR, because several regions of
Leptospira genome are known to possess base
sequences complementary to it11.

Laboratory Manual 55
PCR is performed in 50 ml reaction volumes Materials and equipment required
with 50 ng puried DNA, 2 mM primer, Tris borate bufferv Boric acid
250 mM of each dNTP, 1.5 mM MgCl2, 0.5 U EDTA, disodium Agarose type II
of Taq polymerase in 10 mM Tris-HCl (pH 9.0) Lambda marker Restriction endonuclease
and 50 mM KCl. The temperature programme and buffers
consisted of 1 cycle of 3 min at 94C, 1 min at Ethidium bromide Triple distilled water
60C, and 2 min at 72C; followed by 38 cycles Electrophoresis apparatus Vortex mixture
of 1 min at 94C, 1 min at 60C, and 2 min at Polaroid camera with stand Magnetic stirrer
72C; and a nal cycle of 1 min at 94C, 1
Magnetic stirrer Water bath shaker
min at 60C, and 9 min at 72C. All reaction
Micropipettes & Microtips Gel casting platform with
products are electrophoressed on 20 cm long
comb
1% agarose gels, stained with 0.5 mg/ml
Gel documentation system
ethidium bromide, and photographed under
ultraviolet light using gel documentation
Preparation of buffers
system. DNA proles of individual lanes are
Tris-borate buffer (TBE buffer) pH. 8.2
matched with each other and dendrograms
stock solution (10 x)
of genetic distance based on similarity-
Tris 108.0 gm
coefcient are generated at 5% condence
Boric acid 55.0 gm
level using included software.
EDTA, disodium 9.3 gm
To one litre triple distilled water
RESTRICTION ENDONUCLEASE
ANALYSIS (REA) Working solution 1 x TBE buffer
Stock solution of TBE buffer 100 ml is mixed
properly with 900 ml of triple distilled water to
Principle
prepare the working solution of 1 x TBE buffer.
The Restriction Endonuclease Analysis (REA)
is based upon the fact that DNA of different
Gel loading buffer
nucleotide sequences, those which have a
Bromophenol blue 0.25 gm
different genetic origin will give different
Xylene cyanol 0.25 gm
band pattern in gel electrophoresis after
Sucrose 40.00 gm
enzymatic treatment. The REA involves
Single strength Tris borate
the extraction of DNA from a homogeneous
buffer to 100 ml
population of organisms, digestion of the
DNA with the restriction endonuclease and
Procedure
electrophoresis of the digested DNA in an
The REA is carried out as per the method of
agarose gel. The restriction endonuclease
Marshall et al12. The restriction digestion
recognize and cleave double stranded DNA at
is carried out in 20 l of volume in a sterile
a specic sequence, hence a set of fragments
microfuge tube, the reaction mixture contains
are generated. These fragments are highly
5 l of DNA, 10 x RE buffer 2 l, 10 to 15 units
specic for each type of leptospires for their
restriction enzymes and triple distilled water
molecular characterization. The REA technique
to a nal volume of 20 l.
is sensitive enough to differentiate between
different leptospiral strains on the basis of The digestion mixture is incubated in a water
genetic differences. By comparing restriction bath-shaker at 37C for the required period of
map of different strains, it is possible to time. After the incubation the reaction mixture
differentiate them at molecular level. The (sample) is inactivated by heating at 65C for 5
technique is very much useful for the molecular min before electrophoresis. For electrophoresis
identication/typing of leptospiral isolates/ 0.7% agarose gel in TBE buffer containing 5 g/
strains from various sources. ml ethidium bromide is used.

56 Leptospirosis
Each digested DNA samples are mixed with 1/5th sources and for epidemiological investigations
volume of the gel loading buffer and loaded in in the spread of particular strains in the
the wells of agarose gel. Lambda DNA digested community.
with either Pst I, EcoRI, or Hind III are used as
DNA molecular weight markers. Preparation of buffers
Tris-borate buffer (TBE buffer) pH. 8.2 stock
Electrophoresis is carried out in a sub-marine solution (10 x)
electrophoresis apparatus at 5 v/cm at room Tris 108.0 gm
temperature for 5 to 6 hours, depending upon Boric acid 55.0 gm
the length of the gel. EDTA, disodium 9.3 gm
Triple distilled water to 1.0 litre
At the end of the electrophoresis the gel
is visualized under UV transilluminator and Working solution 1 x TBE buffer
photograph is taken using gel documentation Stock solution of TBE buffer 100 ml is mixed
system. with 900 ml of triple distilled water to prepare
the working solution of 1 x TBE buffer.
RESTRICTION FRAGMENT LENGTH
POLYMORPHISM (RFLP) Gel loading buffer
Bromophenol blue 0.25 gm
Principle Xylene cyanol 0.25 gm
The Restriction Fragment Length Sucrose 40.00 gm
Polymorphism (RFLP) technique is a DNA Single strength Tris borate
based technique which was introduced buffer to 100 ml
in 1990 as a powerful tool for detecting
differences among the chromosomes of 20 X SSC (pH 7.0)
organisms that are very closely related as Sodium chloride (3.0 M) 175.32 gm
determined by the conventional genomic Trisodium citrate (0.3 M) 88.20 gm
analysis. By RFLP analysis the difference in to 1 litre distilled water
DNA sequences are determined by examining 5. 0.25 N HCl
the size of the fragments that result when 20.8 ml of conc. Hcl diluted to a litre with
DNA is cleaved with restriction enzymes. distilled water
RFLP analysis involves the extraction of DNA
from a homogeneous population of organisms, Denaturation solution
digestion of the DNA with the restriction NaCl (1.5 M) 87.76 gm
enzymes and electrophoresis of the digested NaOH (0.5 M) 20.09 gm
DNA in an agarose gel, southern blotting to 1 litre distilled water stored at room
on to nitrocellulose/nylon membranes and temperature
hybridization with specic probes.
Neutralization solution (pH 7.2)
The different restriction enzymes have NaCl (1.5 M) 87.76 gm
different recognition sites and cleave the Tris - HCl (0.5 M) 60.55 gm
DNA at a specific sequence, which produce to 1 litre distilled water
different banding patterns called as DNA
fingerprints. It is now feasible to characterize Prehybridization and hybridization buffer
strains of leptospires/organisms at the Ficoll 400 0.5 gm
genomic level by the unique restriction Bovine serum albumin 0.5 gm
fragment length polymorphism patterns of Polyvinyl pyrralidone 0.5 gm
their DNA. Therefore this technique is very Sodium dodecyl sulphate 2.5 gm
much useful for the molecular identification 20X SSC 125 ml
of leptospiral isolates/strains from various 1M Tris/HCl (pH 7.5) 25 ml

Laboratory Manual 57
0.5M EDTA 1 ml in the agarose gel. Lambda DNA digested with
Deionized formamide 250 ml either EcoRI, Hind III or Pst I can be used as a
DNA molecular weight markers.
Make the volume up to 500 ml with sterilized
triple distilled water. Electrophoresis is carried out in a submarine
electrophoresis apparatus at 5 V/cm at
Procedure room temperature for 5 to 6 hours or 3 V/cm
The restriction digestion is carried out in overnight. At the end of the electrophoresis the
20 l of volume in a sterile microfuge tube, the gel is visualized under UV transilluminator to
reaction mixture contains 10 l of DNA, 10 X RE see the positions of the bands, after that acidic,
buffer 4 l, 3 l restriction enzyme and triple alkaline and neutralization treatment is given to
distilled water to a nal volume of 20 l. the gel before transfer onto nitrocellulose/nylon
membrane, to keep the DNA strands separate.
The reaction mixture is incubated in a water
bath shaker at 37C overnight for complete Southern blotting is done onto nitrocellulose
digestion. After incubation the reaction membrane using transfer buffer for 20-
mixture (sample) is inactivated by heating at 24 hours as described by Southern (1975).
65C for ve min before electrophoresis. For After blotting the membrane is dried at
electrophoresis 1% to 2% agarose gel in TBE room temperature and baked at 80C for
buffer containing 5 g/ml ethidium bromide is two hours in a vacuum oven to x the DNA to
used. the nitrocellulose membrane.

Each digested DNA sample is mixed with 1/5th The nitrocellulose membrane is placed in a
volume of the gel loading buffer and loaded hybridization tube with hybridization buffer (10

Materials and equipment required


Tris borate buffer (pH 8.2 20 X SSC (pH 7)
Sodium saline citrate Sodium chloride
Sodium hydroxide Hydrochloric acid

Sulphuric acid EDTA


Ethidium bromide Agarose type II
Denaturation solution Neutralization solution
Hybridization solution Triple distilled water
Restriction enzymes with appropriate buffers Lambda DNA marker
y-p or DNG labelling kit for specic probe labeling & ECL Random primed labeling kit
detection kit
Nitrocellulose/Nylon membranes Whatmans No. 3 lter paper
Probe DNA can either be a cloned DNA fragment of leptospiral DNA
DNA rRNA
a PCR product eluted from the gel a symthetic oligonucleotide having complementary
sequence to leptospiral DNA
Electrophoresis apparatus UV Transilluminator
Water bath shaker magneti stirrer
Micropipettes & Microtips Hybridization oven
-20C and -70C Vacuum blotter
Vortex mixture Gel casting platform with comb
X-ray lm & Auto-radiography cassettes with intensifying
screens

58 Leptospirosis
to 15 ml) for pre-hybridization at 60C for two overnight to X-ray lm in an auto-radiographic
hours. Then specic probes (DNA, rRNA, cDNA & cassettes with intensifying screen at -70C in
synthetic oligonucleotide having complementary the case of radioactive probe. After enough
sequence to leptospiral DNA) labelled either by exposure the X-ray lm is removed from the
P32 or with digoxigenin, ECL based non-radio cassette and developed.
active method is added to the pre-hybridized
membrane in hybridization solution. In the case of non-radioactive labeling detection
system, the membrane is directly treated with
The hybridization is carried out and incubated ECL luminescent detection buffer/system. If
at 60C for 18 to 24 hours. After hybridization the samples have complementary nucleic acid
the nitrocellulose membrane is washed twice sequences to the probe, the strong bands of
for 15 min in 2 X SSC, once for 15 min in 0.1 restriction fragments are seen/detected on
X SSC at 50C. After washing the membrane the X-ray lm/Nitrocellulose membrane within
is dried at room temperature and exposed 15-30 min.

Laboratory Manual 59
References
1. Millar BD, Chappel RJ, Alder B. 1987. Detection of leptospires in biological uids using DNA hybridization. Vet
Microbiol. 15: 71-78.
2. Terpstra WJ, Schoone GJ, Lighthart GS, Schegget JT. 1987. Detection of Leptospira interrogans in clinical specimens
by in situ hybridization using biotin labeled DNA probes. J Gen Microbiol 133: 911-914.
3. Gerritsen MJ, Olyhoek T, Smits MA, Bokhout BA. 1991. Sample preparation method from polymerase chain reaction-
based semi quantitative detection of Leptospira interrogans, serovar hardjo serotype Hardjo bovis in bovine urine. J
Clin Microbiol 29:2805-2808.
4. Gravekamp C, Van de Kemp H, Franzen M, Carrington D, Schoone GJ, Van eys GJJM, Everard COR, Hartskreerl RA,
Terpstra WJ. 1993. Detection of seven species of pathogenic leptospires by PCR using two sets of primers J Gen
Microbiol. 139: 1691-1700.
5. Woo THS, Patel BKC, Smythe LD, Symonds ML, Norris MA, Dohnt MF. 1997. Comparison of two PCR methods for rapid
identication of Leptospira genospecies Interrogans. FEMS Microbiol Lett 155: 169-177.
6. Terpstra WJ, Schoone GJ, Ter Schegget J. 1986. Detection of Leptospiral DNA by nucleic acid hybridization with 32P
and biotin labeled probe. J Med Microbiol 22:23-28.
7. Van Eys GJJM, Gravekemp C, Gerritsen MJ, Quint W, Cornelissen MTE, Schegget JT, Terpstra WJ. 1989. Detection of
leptospires in urine by the polymerase chain reaction. J Clin Microbiol 27: 2258-2262.
8. Merien F, Perolat P, Maneel E, Persan D, Baranton G. 1993. Detection of Leptospiral DNA by polymerase chain reaction
in aqueous humor of a patient with unilateral uveitis. J Infect Dis 168: 1335-1336.
9. Boom R, Sol CJA, Salimans MMM, Jansen CL, Wertheim-Van Dillen PME & Van der Noorda J (1990) Rapid and simple
method for purication of nucleic acids. J Clin Microbiol 28, 495-503.
10. Roy, S., Biswas, D., Vijayachari, P., Sugunan, A.P., and Sehgal, S.C. (2004). A 22-mer primer enhances discriminatory
power of AP-PCR ngerprinting technique in characterization of leptospires. Tropical Medicine & International Health
9, 1203-1209.
11. Zuerner RL & Bolin CA (1997) Differentiation of Leptopsira interrogans isolates by IS1500 hybridization and PCR
assays. J Clin Microbiol 35, 2612-2617.
12. Marshall RB, Wilton BE, Robinson AJ. 1981. Identication of Leptospira serovars by restriction endonuclease analysis.
J Med Microbiol. 14: 163-166.

60 Leptospirosis
Annexures

Preparation of EMJH Medium


ANNEXURE I

Glassware Take necessary stock solutions out of the


Conical asks freezer.
Measuring cylinders Add 1.5 ml Calcium Chloride and Magnesium
Pipettes Chloride stock solution.
Chemicals required Add 1 ml Zinc Sulphate stock solution.
Na2 HPO4 KH2 PO4
NaCl NH4Cl
Add 100 l Copper Sulphate stock solution.
Thiamine Glycerol Add 50 mg Ferrous Sulphate and 40 mg
Bovine Serum Albumin Fraction V Sodium Pyruvate Sodium Pyruvate.
CaCl2 2H2O MgCl2 6 H2O Add 1 ml of Vit.B12 Stock solution.
FeSO4 7H2O CuSO4 5H2O
ZnSO4 7H2O Vit. B12
Add 12.5 ml Tween 80 stock solution.
Tween 80 Add 24 ml Sterile distilled water to make up
nal volume of 100 ml.
Stock solutions Adjust pH to 7.4 - 7.6 with 1 N NaOH.
Albumin fatty acid supplement stock Filter through using a Millipore lter pore
solution size 0.22 m.
Ingredients
Reagents Grams per Storage Basal Medium
100 ml H2O Dissolve 1 gm of Na2HPO4 and 300 mg of
CaCl2 2H2O + MgCl2 6H20 1.0 each - 20C KH2PO4 and 1 gm of NaCl in 100 ml glass
ZnSO4 7H2O 0.4 - 20C distilled water.
CuSO4 5H20 0.3 + 4C Add 1 ml Ammonium Chloride stock solution.
Vitamin B12 0.02 - 20C
Add 1 ml Thiamine stock solution.
Tween 80 10 - 20C
Add 1 ml Glycerol stock solution.
Basal medium stock solution Add glass distilled water to make up a total
Ingredients vol. of 900 ml.
Reagents Grams per 100 Storage
Adjust pH to 7.4.
ml H2O
NH4Cl 25.0 - 20C Finally make the volume to 1000 ml by
Thiamine 0.5 - 20C adding glass distilled water.
Glycerol 10 - 20C Autoclave at 121C for 30 min.

Method of Preparation Preparation of Complete Medium


Ingredient Parts Quantity
Albumin Fatty Acid Supplement Basal Medium 9 900 ml
Dissolve 10 gms of bovine serum albumin in Albumin Fatty Acid Supplement 1 100 ml
60 ml. sterile glass distilled water. Total 1000 ml

Laboratory Manual 61
Preparation of Fletchers Medium
ANNEXURE II

Reagents required Add 80.8 ml. Sorensen Buffer A and


19.2 ml Sorensen Buffer B.
Na2HPO4 KH2PO4
Mix thoroughly.
NaCl Bacto peptone
Adjust the pH to 7.4 - 7.6.
Bacto Beef Extract Agar
Heat until all ingredients are dissolved.
Stock Solutions Dispense the medium in bottles, 184 ml per
Sorensen Buffer A: Dissolve 11.876 gms. Na2HPO4 bottle.
in one litre of distilled water.
Autoclave all the bottles at 121C for
30 min.
Sorensen Buffer B: Dissolve 9.078 gms. KH2PO4
in one litre distilled water. Store at 4C.

Both Buffer solutions are autoclaved at 121C Preparation of nal medium


for 30 min and stored at 4C. Collect rabbit serum with a little bit of red
blood cells.
Method of Preparation Inactivate the serum for 30 min at 56C in a
Dissolve the following reagents in 820 ml water bath.
distilled water
Add 16 ml of serum to 184 ml medium.
a) Bacto Peptone 0.3 gms.
Dispense the medium in 4 to 5 ml volume in
b) Sodium Chloride 0.5 gms. screw capped test tubes.
c) Bacto Beef Extract 0.2 gms. Check for sterility by incubating the tube
d) Agar 1.5 gms. for 48 hrs at 37C.
Store the medium at 4C until use.

62 Leptospirosis
Maintenance of Leptospires
ANNEXURE III

Routine cultures Stock Culture


Stains used routinely as sources of antigens Stock cultures are best maintained in tubes of
for serological tests or other purposes are semi-solid media and stored in dark at room
maintained in liquid medium dispensed in 5 ml temperature. Cultures are viable for at least
amounts in screw-capped test tubes. Duplicate three months and frequently as long as one
tubes should be inoculated. The inoculum should year. The process of sub-culture is the same
be approximately 10% of the culture volume and as above.
should be examined microscopically to conrm
the presence of viable organisms and the Leptospires cannot readily be stored in frozen
absence of contamination. Cultures are kept at dried state but may be stored for years in
room temperature after incubation of 5 to 7 days liquid nitrogen by using either glycerol
at 30C. Cultures are routinely transferred at (5 to 10%) or di-methyl sulfoxide (5%) as a
2 to 3 weeks intervals. cryoprotectant.

Laboratory Manual 63
Purication of Contaminated Cultures
ANNEXURE IV

Filtration Plating on solid media


Leptospires can pass through a cellulose Leptospiral colonies spread horizontally
membrane lter with pore diameter of through media containing 1% of agarose,
0.22 m. The contaminated uid culture may away from the immobile colonies of the
be diluted and ltered through a suitable lter contaminating bacteria.
and can be cultured from the ltrate.
Animal inoculation
Selective media Inject the contaminated culture in a hamster
Lightly contaminated cultures may be sub- or a guinea pig intra-peritoneally. Take blood
cultured into a media containing 5-uro-uracil by heart puncture after 10 to 15 min and
(100 g/ml). inoculate the blood in culture medium.

64 Leptospirosis
Preparation of Antisera
ANNEXURE V

Selection of Animals 14 days after the last injection, a small


Young healthy rabbits weighing approximately sample of blood should be taken from the
3 to 5 kgs should be selected. ear vein for testing.
If the homologous titre of serum is between
The rabbits should be pre-tested for leptospira 1:10,000 or above the rabbit should be bled
antibodies prior to inoculation. by heart puncture.

Preparation of Antigens If the titre of the serum is below 1:10,000,


Isolates or reference strains should be cloned a further booster injection of 6 ml of live
by culturing on solid media containing 1% culture is given.
agarose. At least two rabbits should be used for the
preparation of each antiserum.
The antigen is prepared by inoculating a single
colony into EMJH liquid medium. Storage of sera
Dispense the sera in small amounts, into
Procedure the sterile ampoules. Lyophilize and store
Rabbits should be inoculated intravenously at 4C.
into marginal ear veins with successive doses Dispense the sera in small amounts (e.g.
of 5 to 7 days old live culture containing 2 ml) into the sterile ampoules and store at
approximately 2x108 leptospires/ml -70C or -20C
Five injections of 1 ml, 2 ml, 4 ml, 6 ml Treat with preservative and store at 4C
and a further 6 ml should be given at (thiomersal 0.02% or phenol 0.3 %).
seven days interval.

Laboratory Manual 65
Requirements for a Leptospirosis Laboratory
ANNEXURE VI

Given below are the minimum basic Staff and skills required
requirements for establishing a small Special laboratory facilities and skills
leptospirosis laboratory capable of performing are required. Leptospires require a lot of
screening and conrmatory tests for the care. Cultures need to be sub-cultured at
diagnosis of leptospirosis. regular intervals and ltered into fresh
media whenever necessary. One needs to be
Culture media well trained or has to employ well trained
The common culture media used are Korthofs technicians, who should be solely working on
medium, Fletchers medium and EMJH medium. leptospirosis.

Equipment Glass Ware & Plastic Ware


Microscope: A good microscope with dark Screw capped test tubes (125 x 15)
eld illumination is essential Conical asks
Hot air oven and Autoclave Pipettes
Centrifuge Pasture pipettes
Micro pipettes Microscopic slides
Balance Racks for test tubes
Refrigerator, freezer
Glass distillation plant
Laminar ow cabinet

66 Leptospirosis
Measurement of the Density of a Leptospira Culture
ANNEXURE VII

The density of leptospira culture can be 2. Counting of leptospires using


determined by: counting chamber (Hawksey or
Burker - Truck).
1. Estimating the number of leptospires
per eld by dark eld microscopy 3. Measure of the Optical density
Grow the leptospira in a liquid media. Measure the OD of formolized antigens by
Add formalin to the culture (nal spectrophotometer using EMJH medium as
concentration 0.5%). blank at 420 nm.

Dilute the culture in a suitable dilution. Culture OD range between 0.052 to 0.1
(Approximately 2-3 X108 leptospires per ml)
Transfer 10 l of diluted culture on a
may be used as an antigen in MAT.
microscopic slide.
Apply cover slip. Using the McFarland scale
Count the leptospires under dark eld The culture density corresponding to that
microscope. of McFarland tube No. 1 (Approximately
Cultures with a minimum density of 600 2-3 x 108 leptospires)
800 leptospires per eld may be used as
antigens.

Laboratory Manual 67
General Safety Rules for Leptospirosis Laboratories
ANNEXURE VIII

1. In connection with the dangers of 2. Make sure the work place is orderly with an
contamination, it is not permitted to easily cleanable surface. Keep the benches
smoke, eat or drink in laboratories, with the and work places as empty as possible. Do
exception of specially allocated places. not sit on worktops. Workplaces and oors
2. All procedures, which could involve must be kept tidy.
chances of direct or indirect contact with 3. Pipetting must never be done with mouth.
the organism or clinical material, are very Use an automatic pipette or a pipette using
hazardous. The use of gloves and the wearing suction-balloon.
of lab coat are thus highly recommended. 4. A clean, disinfected workplace is extremely
3. Hypodermic needles, scalpels, microscopic important in the prevention of contamination.
slides and similar sharp objects should be Make sure to wash your hands with soap and
deposited immediately after use in the water, not only when leaving the workplace,
allocated containers containing disinfectants. but also preferably between procedures.
In order to reduce the chance of prick 5. Working condition is an established
accident, hypodermic needles should never point of attention in the work-assessment
be replaced in the protective sleeves. meetings of the department. Co-workers
4. All the test tubes and vials containing are expected to offer their own points of
organism or patient material should be attention concerning working conditions at
thoroughly closed and should be deposited these meetings. Unsafe situations must be
immediately in the allocated containers immediately reported to their supervisors.
containing disinfectants and should be 6. Proper disposal of clinical specimens,
autoclaved. laboratory waste and chemical waste is of
5. When performing procedures whereby fundamental importance, for both yourself
aerosol formation, splashing or powder and others.
formation can occur, it is mandatory to use 7. In the event of an accident, where staff
protective devices such as safety goggles, are infected or believed to have a risk
face masks, gloves and extractor hoods. of infection, prophylactic medication is
advisable. Precautions should be taken
Safety Recommendations while handling fresh isolates, serum or
1. Keep the laboratory area and administration
blood from the suspected patients.
as far apart as possible

68 Leptospirosis
Typing of Leptospira Isolates Reporting Format
ANNEXURE IX

Determination of serogroup status: Agglutination results with the


standard group sera

Name of the isolate:


Source:

Serogroup Serovar Strain Titre


1. Australis Australis Ballico
2. Autumnalis Rachmati Rachmat
3. Ballum Ballum Mus 127
4. Bataviae Bataviae Swart
5. Canicola Canicola H. Uterecht IV
6. Celledoni Cellodoni Cellodoni
7. Cynopteri Cynopteri 3522 C
8. Dejasiman Dejasiman Dejasinman
9. Grippotyphosa Grippotyphosa Moskva V
10. Hebdomadis Hebdomadis Hebdomadis
11. Icterohaemorrhagiae Icterohaemorrhagiae RGA
12. Javanica Poi Poi
13. Louisiana Louisiana LSU 1945
14. Manhao Manhao L 60
15. Mini Mini Sari
16. Panama Panama CZ 214 K
17. Pomona Pomona Pomona
18. Pyrogenes Pyrogenes Salinem
19. Sejroe Hardjo Hardjopraj
20. Shermani Shermani 1342 K
21. Sermin Weaveri CZ390

Laboratory Manual 69
Leptospirosis

Laboratory Manual

Regional Medical Research Centre


Indian Council of Medical Research
Port Blair

Anda mungkin juga menyukai