Next Article in Journal
Organ Damage and Hepatic Lipid Accumulation in Carp (Cyprinus carpio L.) after Feed-Borne Exposure to the Mycotoxin, Deoxynivalenol (DON)
Next Article in Special Issue
The Effects of the Toxic Cyanobacterium Limnothrix (Strain AC0243) on Bufo marinus Larvae
Previous Article in Journal
Light Influences How the Fungal Toxin Deoxynivalenol Affects Plant Cell Death and Defense Responses
Previous Article in Special Issue
Food Poisonings by Ingestion of Cyprinid Fish
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tetrodotoxin: Chemistry, Toxicity, Source, Distribution and Detection

1
Mass Spectrometry Research Centre (MSRC) and PROTEOBIO Research Groups, Department of Chemistry, Cork Institute of Technology, Rossa Avenue, Bishopstown, Cork, Ireland
2
Department of Chemistry, University of Pune, Pune 411007, India
3
Nanotechnology Group, Tyndall National Institute, University College Cork, Lee Maltings, Cork, Ireland
*
Author to whom correspondence should be addressed.
Toxins 2014, 6(2), 693-755; https://doi.org/10.3390/toxins6020693
Submission received: 20 November 2013 / Revised: 24 January 2014 / Accepted: 26 January 2014 / Published: 21 February 2014
(This article belongs to the Collection Marine and Freshwater Toxins)

Abstract

:
Tetrodotoxin (TTX) is a naturally occurring toxin that has been responsible for human intoxications and fatalities. Its usual route of toxicity is via the ingestion of contaminated puffer fish which are a culinary delicacy, especially in Japan. TTX was believed to be confined to regions of South East Asia, but recent studies have demonstrated that the toxin has spread to regions in the Pacific and the Mediterranean. There is no known antidote to TTX which is a powerful sodium channel inhibitor. This review aims to collect pertinent information available to date on TTX and its analogues with a special emphasis on the structure, aetiology, distribution, effects and the analytical methods employed for its detection.

1. Introduction

In Japan fugu or puffer fish, is a long established delicacy, in spite of its known potential for toxicity. Tetrodotoxin (TTX) is the naturally occurring toxin that is mainly responsible for the risk to consumers [1,2,3,4,5,6,7,8,9,10,11,12,13]. In fact, over 20 species of puffer fish have been found to harbour the toxin [14]. TTX is both water soluble and heat stable so cooking does not negate its toxicity; rather it increases toxic effect [15]. Consequently, great care must be taken by specially trained chefs to ensure that the hazardous parts of the fish (specifically the liver, ovaries and skin) are excised before serving; nevertheless human intoxications and some fatalities have been linked to the consumption of puffer fish [1,2,3,6,9,11,16,17,18,19].
The toxin was first discovered in 1909 by Dr. Yoshizumi Tahara from the ovaries of globefish [20], but puffer fish have been known to be toxic to humans for a long time. TTX is a very potent neurotoxin that is found in a variety of marine [3,5,10,21,22,23,24,25,26,27,28,29] and also in some terrestrial [30,31,32,33,34,35,36,37] species. Its toxicity is often emphasised by referring to the fact that it is over a thousand times more toxic to humans than cyanide; TTX has no known antidote [38,39].
Besides puffer fish, other species known to harbour TTX include: gastropods [4,5,7,8,10,12,13,25,28,40,41,42,43,44,45,46,47,48], newts [31,32,35,36,37,49,50,51,52,53,54,55,56,57,58,59,60], crabs [61,62,63,64,65,66,67,68,69], frogs [30,33,34,70,71,72], sea slugs [73,74], star fishes [75], blue-ringed octopuses [76,77,78], ribbon worms [22,79] and bacteria [74,80,81,82,83,84,85,86,87,88]. The distribution of TTX and its analogues are known to be organism and/or tissue specific [46].
There is some conjecture as to the origin of TTX in puffer fish. It has been shown that cultured non-toxic puffer fish when fed on a diet containing TTX become toxic [89] and toxic puffer fish when fed on a TTX-free diet become non-toxic [90]. It has also been demonstrated that the source of TTX in puffer fish is an endo-symbiotic bacteria that naturally inhabits the gut of the animal [81,82,87,88,91,92]. This may be explained by the hypothesis that puffer fish could initially acquire the TTX producing bacteria via the food web and that these bacteria then persist in the fish [93]. Indeed several ubiquitous varieties of bacteria produce TTX including some in the Pseudomonas and Vibrio genera [21,80,81,82,83,84,85,86,87,88,91,92,94,95]. Origin of TTX in terrestrial animal’s, for example newt and frog is endogenous because it has role in defense from predators in these animals (Refer to paragraph 4.3).
The mechanism of TTX toxicity has been investigated in animal models [15,96,97]: It is a sodium channel blocker. The toxin binds to the sodium channels of the excitable tissues of the victim (muscles and nerves); the inhibition of sodium ions through the channels effectively immobilises these tissues [98]. In humans the onset and severity of the symptoms of TTX poisoning after ingestion is dose dependent [19]. Initial symptoms include tingling (paresthesias) of the tongue and lips, followed by or concurrent with headache and vomiting, which may progress to muscle weakness and ataxia. In severe cases death may occur due to respiratory and/or heart failure [6]. The only treatment for TTX intoxication is observation and appropriate supportive care [38].
TTX was regarded until the recent past as a problem confined to Japan and Asian countries; now the problem is emerging as a threat to regions that were here to fore considered safe, refer to Table 1. It is thought that the “spread” of the toxin is due to increasing water temperatures world-wide [99]. In this review we will examine reports regarding the geographic distribution of TTX (Table 1).
Table 1. Worldwide occurrence of tetrodotoxin (TTX) poisoning.
Table 1. Worldwide occurrence of tetrodotoxin (TTX) poisoning.
CountryCausative OrganismAnalogues of TTX foundNo. of casesYear of poisoning incidentReference
AustraliaToadfishTTX72004[100]
AustraliaPuffer fishTTX111 January 2001 to 13 April 2002[101]
BangladeshPuffer fish, Dora potka i.e., Takifugu oblongus in Natore District and Badami potka i.e., Arothron stellatus in Narsingdi DistrictTTX141 (Three outbreaks)
48 (Narsingdi District) + 10 (Dhaka) + 83 (Natore District)
9 April 2008 (Narsingdi District)
3 June 2008 (Dhaka)
8 June 2008 (Natore District)
[19]
BangladeshPuffer fishTTX53May 2001–May 2006[102]
Bangladesh (Khulna district)Puffer fishTTX37 (8 died)April 2002[9]
Bangladesh (Degholia in the Khulna district)Puffer fish, Takifugu oblongusTTX36 (7 died)May 2002[11]
BangladeshPuffer fish, Takifugu oblongusTTX8 (5 died)1998[3]
China (Lianyungang)Gastropod, Nassarius spp.TTX, trideoxyTTX, 4-epiTTX, anhydroTTX and oxoTTX-May–August 2007[28]
China (South Zheijiang, Mainland)Gastropod, Zeuxis samiplicutusTTX30June 2001[5]
India (Orissa state, Burla)Puffer fishTTX8October 2007[17]
JapanTTXNumerous1965–2010[46]
JapanPuffer fish “kinfugu”, T. poecilonotusTTX1October 2008[103]
JapanThread-sail filefish “Kawahagi”TTX1October 2008[103]
JapanMarine snail, Nassarius glansTTX1July 2007[103]
JapanMarine snail, C. saulieTTX2December 1987[46]
JapanMarine snail, C. saulieTTX1December 1979[40]
JapanMarine snail (Ivory shell), Babylonia japonicaTTX5June 1957[16]
KoreaUnknown fishTTX3October 2010[104]
Mediterranean region (Egypt and Israel)Puffer fish, L. sceleratusTTX13
(9) Hafia bay, (2) Caesarea coast, (2) Ashkelon coast
November 2005, February 2007, March 2007, November 2007, March 2008 and May 2008[105]
New ZealandGrey side-gilled sea slug, Pleurobranchaea maculataTTX15 dogsJuly to November 2009[46]
Spain (Malaga; caught from Portuguese waters)Trumpet shell, Choronia lampusTTXOctober 2007[45]
Taiwan (Kaohsiung)Gastropod, Niotha clathrataTTX3November 2006[13]
TaiwanGastropodTTX and PSP1October 2005[12]
Taiwan (Southern China Sea)Marine snail,
Nassarius (Alectricon) glans
TTX5April 2004[46]
Taiwan (Tungsa Island)Gastropod, Nassarius glansTTX6April 2004[10]
Taiwan (Western)Gastropod, Polinices didyma and Natica lineataTTX2003[8]
Taiwan (Tungkang, Southern Taiwan)Gastropods, Oliva miniacea, Oliva mustelina and Oliva niraseiTTX1February 2002[7]
TaiwanUnknown fishTTX6 (1 died)April 2001[106]
TaiwanPuffer fish, Lagocephalus lunarisTTX6 (1 died)April 2001[6]
Taiwan (Northern)Gastropods (snails), Zeuxis sufflatus and Niotha clathrataTTX4April 2001[4]
Taiwan (Chunghua Prefecture, Western Taiwan)Puffer fish, Takifugu niphoblesTTX5Jan 2000[107]
Thailand (Chon Buri, Eastern Thailand)Eggs of horseshoe crab, Carcinoscorpius rotundicaudaTTX711995[63]
US (New Hampshire, New York, Pennsylvania and Virginia)Newt, N. viridescensTTX, 6-epiTTX and 11-oxoTTXCollected samples for analysis2001–2009[37]
US (Chicago)Puffer fishTTX2May 2007[108]
US (California)Puffer fish transported from JapanTTX3April 1996[109]
US (Hawaii)Puffer fish, Diodon hystrixTTX11986[110]
Structurally TTX consists of a guanidinium moiety connected to a highly oxygenated carbon skeleton that possesses a 2,4-dioxaadamantane portion containing five hydroxyl groups [111]. TTX co-exists with its naturally occurring analogues. There have been 30 structural analogues of TTX reported to date (Figure 1) and the degree of toxicity varies with structure [112]. One of the major problems for studying these analogues is the lack of commercially available standards. A number of researcher groups have synthesized some of the analogues of TTX on a laboratory scale [113,114,115,116,117,118,119,120] but availability is severely limited.
Several analytical methods have been used for the detection and quantitation of TTX and its analogues. Mouse bioassay is commonly used for TTX analysis detecting TTX [27,121,122,123] and some of its toxic analogues (5,6,11-trideoxyTTX [124], 11-deoxyTTX [31] and 6,11-dideoxyTTX [125]). Liquid chromatography hyphenated with mass spectrometry (LC-MS/MS) detection is the preferred method for analysis of TTX, due to its unrivalled selectivity [13,25,106,126,127,128]. Toxicity limits for TTX in mice had been established [31,125,129,130], but there is still a need to establish accurate regulatory limits for TTX in humans [46,131]. However, the shortage of standards for the TTX analogues is the main hindrance for such studies to be undertaken.
Figure 1. Structures of TTX analogues.
Figure 1. Structures of TTX analogues.
Toxins 06 00693 g001
It is important to analyse TTX analogues for toxicity because extensive toxicity studies have not been conducted on all the analogues. It is known that the deoxy analogues of TTX are less toxic than TTX, but the hydroxyl analogues are more toxic than TTX, this is likely to have implications in risk analysis studies concerning the effect of the toxin family on human health, please refer to Section 9 for a more in-depth discussion.

2. Structures and Standards for TTX and Its Analogues

The name TTX was coined by Dr. Yoshizumi Tahara in 1909. He isolated TTX from the ovaries of globefish [132]. TTX is a heterocyclic, organic perhydroquinozolineamine molecule (aminoperhydroquinazolone). Its structure was elucidated by R. B. Woodward in 1964 [133]. Authentic standards for TTX are available from various commercial sources (Table 2); Wako Chemicals (Purity >90%) [134], Latoxan (Valence, France) [43], Sigma Aldrich (St. Louis, MO, USA), 99% HPLC [104,135], Sankyo Co., Ltd, Tokyo, Japan [136], Calbiochem, EMD Millipore, USA, [51], Ascent Scientific [137] and Axxora, Grunberg, Germany [138].
All of these suppliers obtain TTX from TTX containing organisms (most commonly puffer fish ovary). These standards sometimes contain other analogues of TTX depending upon the source organism. O’Leary et al., 2004 stated that they had found two peaks in LC-MS analysis of standards for TTX supplied by Sigma-Aldrich, Australia as well as with Latoxan standards [120]. They assumed that the minor peak is anhydro-TTX. Therefore most standards are certified only for TTX (except for Wako Chemicals who supply standards for TTX, 4-epiTTX and anhydroTTX [139]). Though other analogues are present in the vast majority of standards they cannot be accurately quantified against a certified material standard, as there is a lack of analogue standards.
There are 26 naturally occurring analogues of TTX (Table 3). As the authentic standards for these analogues are not commercially available, they have to be extracted and isolated from available sources (with further chemical modification in some cases); TTX was isolated from the ribbon worm, Cephalothrix simula (formerly Cephalothrix sp.) along with 4-epiTTX and 4,9-anhydroTTX [22,40,141]; TTX from the ovaries and the livers of puffer fish, Fugu Pardalis [24]; 11-deoxy TTX from the newt, Cynops ensicauda [31]; TTX, 4-epiTTX and 4,9-anhydroTTX from the eggs of the puffer fishes, Fugu poecilonotus and Fugu pardalis [142]; 5,6,11-TrideoxyTTX from the eggs of F. poecilonotus [124]; 6-epiTTX and 11-deoxyTTX from the newt, Cynops ensicauda [31]; Chiriquitoxin from the frog, Atelopus chiriquiensis [143]; TTX, 4-epi TTX and anhydro TTX from the liver of puffer fish, Takifugu oblongus [8]; TTX, 4-epi TTX, 4,9-anhydro TTX and 5,6,11-trideoxy TTX from the puffer fishes, Fugu poecilonotus [124] and Fugu Pardalis [124]; TTX (94.3% purity) from the liver of puffer, Takifugu oblongus by the method of Hwang et al., 1988 [106,144]; 6-epi TTX and 11-deoxy TTX from the newt, Cynops ensicauda [31] and TTX, 6-epi TTX, 4-epi TTX and 4,9-anhydro TTX from the puffer fish, Spheroids Spengler by the method of Goto et al., 1965 [145]. 11-norTTX-6(S)-ol, 11-norTTX-6(R)-ol and 11-norTTX-6,6-diol have been synthesized [146,147,148,149].
Yotsu-Yamashita et al., 1999 synthesized eight derivatives of TTX by chemical modifications of TTX extracted from the puffer fishes, Fugu poecilonotus and Fugu pardalis: 11-oxoTTX, TTX-11-carboxylic acid, 11-norTTX-6,6-diol, 11-norTTX-6(S)-ol, 11-norTTX-6(R)-ol, tetrodonic acid, 4,9-anhydro-8-O-hemisaccinate and TTX-8-O-hemisaccinate. Though these were just one or two step chemical reactions, the percentage yields of each derivative were 5%, 7%, 73%, 14%, 4%, 75%, 40% and 34% respectively [112].
Four new analogues, 8-epi-5,6,11-trideoxyTTX, 4,9-anhydro-8-epi-5,6,11-trideoxyTTX, 1-hydroxy-8-epi-5,6,11-trideoxyTTX and 1-hydroxy-4,4a-anhydro-8-epi-5,6,11-trideoxyTTX were recently isolated from newt, Cynops ensicauda popei by Kudo et al., 2012 [59].
Apart from the commercially available standards given, very few of the isolated TTX analogues or derivatives listed, are available in sufficient quantities to allow for the development and implementation of widespread screening by quantitative analytical methods.
Table 2. Commercial sources for TTX.
Table 2. Commercial sources for TTX.
No.Name of TTX analogueSource for extractionPurity (%)CAS No.Contact details
1Tetrodotoxin (citrate free)Fugu fish organs96 (HPLC‚ IR‚ NMR)4368-28-9[150]
2Tetrodotoxin (citrate free)Fugu spp.1004368-28-9[151]
3Tetrodotoxin (citrate free)NMNM4368-28-9[152]
4Tetrodotoxin (citrate free)NMNM4368-28-9[153]
5Tetrodotoxin (MW 328.28)Fugu spp.NM4368-28-9[154]
6Tetrodotoxin (MW 319.27) C11H17N3O8NM≥98 (HPLC)4368-28-9[155]
7Tetrodotoxin (citrate free), C11H17N3O8, MW 319.3 NMNM4368-28-9[156]
8Tetrodotoxin (citrate free), C11H17N3O8, MW 319.28Tetraodon pardalis>984368-28-9[157]
9Tetrodotoxin (citrate free), C11H17N3O8, MW 319.3Fugu spp.≥95 by TLC4368-28-9[158]
10Tetrodotoxin (citrate free), C11H17N3O8, MW 319.2NM>984368-28-9[159]
11Tetrodotoxin citrate, C11H17N3O8, MW 319.27Fugu>9818660-81-6[160]
12Tetrodotoxin citrate, C11H17N3O8, MW 319.27NM>984368-28-9[161]
13Tetrodotoxin (citrate free), C11H17N3O8, MW 319.27NMNM4368-28-9[162]
14Tetrodotoxin (citrate free), C11H17N3O8, MW 319.3NMNM4368-28-9[163]
Note: NM, Not mentioned.
Table 3. Sources of TTX analogues.
Table 3. Sources of TTX analogues.
No.AnalogueExact Molar MassMolecular FormulaMRMSource Organism
14-S-Cys TTX 422.110753C14H22N4O9SNRPuffer fish: Fugu Pardalis [24,164]
2TTX-8-O-hemisuccinate418.109787C15H20N3O11NRSynthetic analogue [112]
3Chiriquitoxin 392.117945C13H20N4O10NRToad: Atelopus chiriquiensis [30,70,143]
411-oxoTTX 335.096482C11H17N3O9336/318
336/300
336/282
336/178
336/162
(i)
Puffer fish: Arothron nigropunctatus [165]
(ii)
Gastropod: Nassarius spp. [28]
(iii)
Xanthid crab: Atergatis floridus [62]
(iv)
Newt: Taricha spp. [36] and Notophthalmus viridescens [36,37,53]
(v)
Frog: Brachycephalus ephippium [34] and B. pernix [72]
5TTX-11-carboxylic acid332.073007C11H14N3O9NRSynthetic analogue [112]
6TTX319.101567C11H17N3O8320/302
320/162
(i)
Puffer fish: F. niphobles [26,166], T. nigroviridis [26], T. biocellatus [26], Lagocephalus sceleratus [29,47,48], L. Lunaris [27], L. Spadiceus [27], Fugu poecilonotus [59], Fugu obscurus [87], Fugu pardalis [24,26,125], Fugu rubripes [82], Fugu niphobles [26], Takifugu oblongus [138], Takifugu vermicularis [23], Takifugu rubripes [90], Arothron immaculatus and Arothron nigropunctatus [148,165], Arothron firmamentum [167], A. reticularis [27], Chelonodon patoca [23], Xenopterus naritus [27], Tetraodon Nigroviridis [26,27], Tetradon biocellatus [26], Sphoeroides annulatus [168], S. Lobatus [168], S. lispus [168], Arothron meleagris [168], Canthigaster punctatissima [168], Takifugu niphobles [169], Pseudocaligus fugu [169] and Taeniacanthus sp. [169]
(ii)
Gastropod: Nassarius spp. [25,28], Gibbula umbilicalis [47], Monodonta lineata [47], Charonia lampas [45,47,48], N. nitidus [25], N. semiplicatus [28], N. papillosus [12], Niotha clathrata [4,13,41,42], Charonia sauliae [46], Nassarius (Alectricon) glans [10,46], Babylonia japonica [46], Zeuxis scalaris [42,46], Zeuxis samiplicutus [5], Z. sufflatus [4,46], Z. siquijorensis [46], Polinices didyma [8], Natica lineata [8], Oliva miniacea [7], O. mustelina [7] and O. nirasei [7].
(iii)
Crab: Demania cultripes [139,170], Xanthias lividus [67,171], Carcinoscorpius rotundicauda [63,172], Demania toxica [139], Demania reynaudi [65,139], Lophozozymus incises [139], Lophozozymus pictor [139], Atergatopsis germaini [139], Atergatis floridus [62,65,139], Zosimus aeneus [171], Actaeodes tomentosus [171] and Camposcia retusa [171].
(iv)
Newt: Taricha spp. [36], Taricha granulosa [32,49,51,54,58], Cynops ensicauda popei [59], Taricha torosa [55,56], Triturus spp. [35], Notophthalmus viridescens [32,36,37,50,53,57,60], Cynops spp. [32], Cynops pyrrhogaster [52], 9Cynops ensicauda [31,173], Triturus alpestris [35], Triturus cristatus [35], Triturus helveticus [35] and Triturus vulgaris [35]
(v)
Frog: Brachycephalus ephippium [33,34,72], B. pernix [72], Polypedates sp. [71] and Atelopid frogs [30]
(vi)
Blue-ringed octopus: Hapalochlaena lunulata [76,77,78], Hapalochlaena fasciata [76,78] and Octopus maculosus [94]
(vii)
Starfish: Astropecten scoparius [75]
(viii)
Sea slug: Pleurobranchea maculata [73,74]
(ix)
Copepod: Pseudocaligus fugu ectoparasitic on the panther puffer Takifugu pardalis [85]
(x)
Ribbon worm: Cephalothrix sp. (Nemertean) [22] and Cephalothrix linearis (Nemertean) [79]
(xi)
Bacteria: Bacillus spp. W-3 from Fugu obscurus [87], Vibrio spp. from xanthid crab Atergatis floridus [61], Vibrio alginolyticus from the starfish Astropecten polyacanthus [174], Aeromonas from Takifugu obscurus [88], Bacillus sp. from Fugu obscurus [87], Bacillus spp. and Actinomycete spp. from Fugu rubripes [82], Microbacterium arabino galactanolyticum [81], Serratia marcescens [81], Vibrio alginolyticus [81] from Chelonodon patoca, Bacillus, Micrococcus, Alcaligenes, Caulobacter and Flavobacterium from fresh water sediment [95], Bacillus, Micrococcus, Acinetobacter, Aeromonas, Alcaligenes, Alteromonas, Flavobacterium, Moraxella, Pseudomonas and Vibrio from deep sea sediment [21], Alteromonas, Bacillus, Pseudomonas and Vibrio spp. from the blue-ringed octopus, Octopus maculosus [94], Roseobacter sp. from the copepod Pseudocaligus fugu, parasite of puffer Takifugu pardalis [85], Copepods, Pseudocaligus fugu and Taeniacanthus sp., parasites of puffer Takifugu niphobles [169], Aeromonas from puffer fish, Takifugu obscures [88], and Vibrio Strain, LM-1 from the puffer, Fugu vermicularis radiatus [91]
74-epiTTX319.101567C11H17N3O8320/302
320/162
(i)
Puffer Fish: F. niphobles [26,166], Takifugu nigroviridis [26], Fugu poecilonotos [59], Fugu pardalis [24,125], Takifugu oblongus [138], Tetraodon nigroviridis, Tetradon biocellatus and Lagocephalus sceleratus [29,47], Sphoeroides annulatus, S. lispus, Arothron meleagris, Canthigaster punctatissima and Pseudocaligus Fugu [169] and A. meleagris [168]
(ii)
Gastropod: Nassarius spp. [28], Nassarius glans [10], Charonia lampas and M. lineata [47] and N. semiplicatus [86]
(iii)
Crab: Demania cultripes [170]
(iv)
Newt: Cynops pyrrhogaster [52], Cynops ensicauda popei [59], Cynops ensicauda [173], Notophthalmus viridescens [50,53,57], Triturus spp. [35] and Taricha granulosa [51]
(v)
Frog: Brachycephalus ephippium [33,34]
(vi)
Blue-ringed octopus: Octopus maculosus [94]
(vii)
Copepods: Pseudocaligus Fugu and Taeniacanthus sp., parasites of puffer Takifugu niphobles [169]
(viii)
Ribbon worm: Cephalothrix spp. [22]
(ix)
Bacteria: Vibrio Strain, LM-1 from the puffer, Fugu vermicularis radiatus [91]
86-epiTTX319.101567C11H17N3O8320/302
320/162
(i)
Puffer Fish: Fugu poecilonotos, (<LOD) [59]
(ii)
Newt: Taricha spp. [36], Cynops ensicauda [173], Cynops ensicauda popei [59], Notophthalmus viridescens [32,36,37,50,53,57,60], Cynops pyrrhogaster [52], Triturus alpestris [35], Triturus cristatus [35], Triturus helveticus [35], Triturus vulgaris [35], Taricha granulosa [32] and Cynops spp. [32]
(iii)
Frog: Brachycephalus ephippium [34,72], B. nodoterga [72] and B. pernix [72]
9Tetrodonic acid319.101567C11H17N3O8NR
(i)
Puffer Fish [127]
(ii)
Newt: Cynops ensicauda [173]
(iii)
Frog: Brachycephalus ephippium [33,34]
1011-norTTX-6,6-diol305.085917C10H15N3O8NRSynthetic analogue [112]
115-deoxyTTX303.106652C11H17N3O7304/286
304/176
(i)
Puffer Fish: Lagocephalus sceleratus [29], F. niphobles [26], T. nigroviridis [26], T. biocellatus [26], puffer Fish [127], Fugu poecilonotus [59] and Fugu pardalis [24,125]
(ii)
Newt: Cynops ensicauda popei (<LOD) [59]
(iii)
Frog: Brachycephalus ephippium, B. nodoterga and B. pernix [72]
1211-deoxyTTX303.106652C11H17N3O7304/286
304/176
(i)
Puffer Fish: F. niphobles [26], Takifugu oblongus [138], T. nigroviridis [26], T. biocellatus [26], Lagocephalus sceleratus [29], Fugu poecilonotos [59] and Fugu pardalis [24,125]
(ii)
Newt: Cynops ensicauda [59,173], Cynops spp. [32], Taricha granulosa [32] and Notophthalmus viridescens [32]
(iii)
Frog: Brachycephalus ephippium, B. nodoterga and B. pernix [72]
131-hydroxy-5,11-dideoxyTTX303.106652C11H17N3O7NRNewt: Taricha granulosa [59]
144,9-anhydro TTX301.091002C11H15N3O7302/256
302/162
(i)
Puffer Fish: F. niphobles [26,166], T. nigroviridis [26], T. biocellatus [26], Fugu poecilonotos [59], puffer Fish [127], Fugu pardalis [24,125], A. meleagris [168], Lagocephalus sceleratus [29,47], Sphoeroides annulatus [168] and S. lispus, [168]
(ii)
Gastropod: Charonia lampas [47]
(iii)
Xanthid crab: Demania cultripes [170]
(iv)
Newt: Cynops ensicauda popei [59], Cynops pyrrhogaster [52], Notophthalmus viridescens [50,53,57], Triturus spp. [35] and Taricha granulosa [51]
(v)
Frog: Brachycephalus ephippium [34]
156-epi-4,9-anhydroTTX301.091002C11H15N3O7302/256
302/162
Newt: Cynops pyrrhogaster [52], Notophthalmus viridescens 53,57 and Triturus spp 35,175
16AnhydroTTX300.083177C11H14N3O7302/256
302/162
(i)
Puffer Fish: Takifugu niphobles [169], Takifugu oblongus [138] and Pseudocaligus Fugu [169]
(ii)
Gastropod: Charonia lampas [47], Nassarius spp. [28], Zeuxis samiplicutus and Nassarius glans [10], Natica lineata [8] and Polinices didyma [8]
(iii)
Crab: Xanthias lividus [67,139] and D. Cultripes [139]
(iv)
Newt: Cynops ensicauda [173]
(v)
Frog: Brachycephalus ephippium [33]
(vi)
Blue-ringed octopus: Octopus maculosus [94]
(vii)
Copepods: Pseudocaligus Fugu and Taeniacanthus sp., parasites of puffer Takifugu niphobles [169]
(viii)
Ribbon worm: Cephalothrix spp. [22]
(ix)
Bacteria: Roseobacter sp. from the copepod Pseudocaligus Fugu, parasite of puffer Takifugu pardalis [85] and Vibrio Strain, LM-1 from the puffer, Fugu vermicularis radiatus [91]
1711-norTTX-6(S)-ol289.091002C10H15N3O7290/272
290/162
(i)
Puffer Fish: Lagocephalus sceleratus [29,47], Fugu poecilonotos [59], puffer Fish [127], Fugu pardalis [125] and A. meleagris [168]
(ii)
Frog: Brachycephalus ephippium [33]
(iii)
Newt: Cynops ensicauda popei (<LOD) [59] and Arothron nigropunctatus [148]
(iv)
Sea slug: Pleurobranchaea maculata [73]
1811-norTTX-6(R)-ol289.091002C10H15N3O7290/272
290/162
(i)
Puffer Fish: Lagocephalus sceleratus [29,47], puffer Fish [127] and Fugu niphobles [147]
(ii)
Crab: Atergatis Floridus [62]
(iii)
Newt: Cynops ensicauda [173]
(iv)
Sea slug: Pleurobranchaea maculata [73]
191-hydroxy-8-epi-5,6,11-trideoxy TTX287.111737C11H17N3O6288/162
(i)
Puffer Fish: Fugu poecilonotos (<LOD) [59]
(ii)
Newt: Cynops ensicauda popei [59]
206,11dideoxyTTX287.111737C11H17N3O6288/224
(i)
Puffer Fish: F. niphobles [26], T. nigroviridis [26], T. biocellatus [26], Fugu niphobles and Fugu poecilonotos [59] and Fugu pardalis [125]
(ii)
Newt: Cynops ensicauda popei (<LOD) [59]
218,11dideoxyTTX287.111737C11H17N3O6NRSynthetic analogue [114,119]
225,6,11-trideoxy TTX271.116822C11H17N3O5272/254
272/162
(i)
Puffer Fish: F. niphobles [26], Takifugu oblongus [138], T. nigroviridis [26], T. biocellatus [26], Lagocephalus sceleratus [29,47], puffer Fish [127] Fugu poecilonotos [59] and Fugu pardalis [24,125]
(ii)
Gastropod: Nassarius spp. [28] and Charonia lampas [47]
238-epi-5,6,11-trideoxy TTX271.116822C11H17N3O5272/162Newt: Cynops ensicauda popei [59]
244-epi-5,6,11-trideoxyTTX271.116822C11H17N3O5NRPuffer fish [127]
251-hydroxy-4,4a-anhydro-8-epi-5,6,11-trideoxyTTX269.101172C11H15N3O5270/162
(i)
Puffer Fish: Fugu poecilonotus (<LOD) [59]
(ii)
Newt: Cynops ensicauda popei [59]
264,9-anhydro-5,6,11-trideoxy TTX253.106257C11H15N3O4NRPuffer fish [59]
274,9-anhydro-8-epi-5,6,11-trideoxy TTX253.106257C11H15N3O4254/162Newt: Cynops ensicauda popei [59]
284,4a-anhydro-5,6,11-trideoxy TTX253.106257 C11H15N3O4NRPuffer fish [59]
294-epi-11-deoxyTTXNRNRNRNewt (Cynops ensicauda) [173]
304,9-anhydro-11-deoxyTTXNRNRNRNewt (Cynops ensicauda) [173]
Note: NR, Not Reported.

3. Chemical Synthesis of TTX and Its Analogues

Several groups have synthesised TTX (Figure 2) and some of its analogues including 8,11-dideoxyTTX and 5,11-dideoxyTTX using glucose as precursor molecule [113,114,115,116,117,118,119,120]. The key intermediate compound for the synthesis of TTX can be obtained either from quinone [113] or carbohydrate [119]. Umezawa et al., 2010 [177] have synthesized 13C-labelled 5,6,11-trideoxytetrodotoxin and 13C-labelled 4-epi-5,6,11-trideoxytetrodotoxin from aldehyde. Adachi et al., 2013 [120] have synthesized (-)-5,11-dideoxytetrodotoxin from the enone. Sato et al., 2012 [119] have done stereo selective synthesis of optically active TTX from d-Glucose. Nishikawa et al., 2003 have done stereo controlled synthesis of 11-deoxyTTX and 8,11-dideoxytetrodotoxin from levoglucosenone [116]. Ohyabu et al., 2003 [117] achieved the asymmetric synthesis of tetrodotoxin from 2-acetoxy-tri-O-acetyl-d-glucal.
However in general, chemical synthesis of TTX involves many complex steps (average no. = 23–67) which generate low yields (0.34%–1.82%) of the target compound [111]. Additionally, in most synthesis it is necessary to develop methods for purification. These complications make most laboratory synthesis approaches for TTX and its analogues unfeasible for commercial scale-up.
Figure 2. Chemical synthesis of TTX from glucose [119] (R = MOM).
Figure 2. Chemical synthesis of TTX from glucose [119] (R = MOM).
Toxins 06 00693 g002

4. Aetiology of TTX

4.1. Biosynthesis of TTX

The biosynthetic origin of TTX in vivo has yet to be agreed. It is supposed that arginine is the precursor moiety for TTX production within the organism (Figure 3) [49].

4.2. Aetiology of TTX among Marine and Fresh Water Organisms

The ecology of marine organisms [178] and terrestrial organisms [179] with respect to TTX have been discussed before. There have been several theories proposed for the formation and bio-transfer of TTX. TTX is believed to bio-accumulate via the marine food chain [178] (Figure 4).
It is known that several species of bacteria and other microorganisms often live within larger marine animals in a supposed or in an established mutually advantageous symbiosis [180,181,182]. The following species of bacteria are known TTX producers and have been isolated from various marine organisms: Vibrio alginolyticus from starfish, Astropecten polyacanthus [174]; Vibrio spp. from the puffer fish, Fugu vermicularis radiates [91]; Aeromonas from puffer fish, Takifugu obscures [88]; Vibrio and Pseudomonas spp. from gastropod, Niotha Clathrata [80]. Chau et al., 2011 [111] have provided a comprehensive account of the distribution of TTX producing bacteria in many organisms.
Figure 3. Proposed biosynthesis of tetrodotoxin from arginine [49,111].
Figure 3. Proposed biosynthesis of tetrodotoxin from arginine [49,111].
Toxins 06 00693 g003
Figure 4. Proposed mechanism of TTX accumulation in marine animals [14].
Figure 4. Proposed mechanism of TTX accumulation in marine animals [14].
Toxins 06 00693 g004
A small gastropod, U. suturale had been isolated from the digestive gland of starfish, A. scoparius is a known progenitor of TTX and anhydroTTX [75]. This suggests an exogenous (external) origin for TTX in this starfish.
An exogenous origin for TTX is also suspected for certain toxic crabs which feed on small gastropods known to contain TTX and on marine sediments containing TTX-positive bacteria (food vectors include: Vibrio VIII from xanthid crab [61]; Vibrio and Pseudomonas spp. from gastropod, Niotha Clathrata [80]; Bacillus, Micrococcus, Aeromonas, Alteromonas, Moraxella, Vibrio and Actinetobacter spp. from deep sea sediment [21] and, Bacillus, Micrococcus, Alcaligenes, Caulobacter and Flavobacterium spp. from fresh water sediment [95]). Therefore it is assumed that crabs obtain TTX from the food chain.
The assumption of an exogenous origin of TTX in fresh water and marine organisms is also supported by other studies. It has been shown that puffer fish, Takifugu rubripes [90] and grey side-gilled sea slugs, Pleurobranchaea maculata [74] become non-toxic when they are fed on a TTX-free diet. Also when puffer fish, Takifugu rubripes were fed with a TTX-containing diet their toxicity increased significantly [14].
Contrarily, Matsumura, 1998 [183] have demonstrated that TTX levels in the embryos of puffer fish, Takifugu niphobles increase until hatching; emphasizing its endogenous (internal) origin. Other evidence for an endogenous origin for TTX in gastropods is that TTX was higher in the muscle than in the digestive gland of the snail N. lineata [184]. In addition, TTX toxicity in the edible gastropod, Polinices didyma was higher in the muscle tissue than in the digestive gland [184]. Of course in the case of the gastropods, it is possible that after initial ingestion of TTX, the toxin could have then migrated and remained in tissue compartments outside of the digestive region.

4.3. Aetiology of TTX among Terrestrial Animals

In newts the levels of TTX and 6-epiTTX are higher in the skin rather than in the liver [51]. The hypothesis here is that TTX is biosynthetically produced by the creature as a defense mechanism to deter and immobilise predators. However, researchers have found that the newt, N. viridescens [37] becomes non-toxic when it is fed a TTX-free diet suggesting, that at least in this case that TTX has an exogenous origin.
Pires Jr. et al., 2005 [72] studied TTX in frogs belonging to the Brachycephalus family, noting that the degree of colouration of the frog can be related to its toxicity. They have found the highest TTX levels in the skin followed by liver and ovaries of frogs belonging to the genus B. ephippium and B. pernix which are bright yellow. However B. nodoterga, which has cryptic colouration, was found to be non-toxic. Interestingly, the infamous “poison dart” frogs belonging to Dendrobatidae family are brightly coloured and are well known for high toxicity, though their toxicity derives not only from TTX. The bright colouration protects these frogs from predators who instinctively avoid highly coloured prey [185]. This strongly suggests toxic frogs naturally synthesise the toxin; however studies have not been conducted to definitively corroborate this.
Toxicity in two snails, on which crabs feed, Polinices didyma and Natica lineata collected from Southern Taiwan were higher (1563 and 2590 MU/specimen respectively) than that of these species collected from Western Taiwan (8–123 and 5–95 MU/specimen respectively) [8]. These studies indicate that there is regional variation in toxicity that subsequently correlates with the toxicity of crabs from these regions. This would imply an exogenous origin of TTX in both these snails and the crabs higher up the food chain.
Despite all of these assumptions, the exact origin and pathway for the synthesis and bio-transfer of TTX is not yet fully known and requires further investigation.

5. Biochemistry

TTX is a sodium channel blocker. Binding of TTX to voltage gated sodium channel results from the interaction between the positively charged guanidine group on the TTX with the negatively charged carboxylate groups on the side chains in the mouth of the sodium channel [98,186,187]. TTX binding prevents diffusion of sodium ions through the sodium channels. This in turn prevents depolarization and propagation of action potentials in nerve cells leading to the loss of sensation. Biotransformation of TTX inside the human or mammalian body is yet to be widely investigated. Some information is available on transformation of TTX and/or its analogues inside the bodies of puffer fish [134] and grey side-gilled sea slugs, Pleurobranchaea maculata [74]. Wood et al., 2012 [74] have shown that degradation/transfer of TTX in the heart tissue is the fastest, while TTX degrades/transfers relatively slowly from the gonads. Additionally, there was a very high level of TTX in eggs. In the puffer fish body, TTX enters the liver first after ingestion. Then it is transferred mainly to the skin in the male and the reproductive organs in the female [103,134]. As discussed in Section 6, TTX binding proteins isolated from marine puffer fish can have a major role in the biotransformation of TTX in the puffer fish body [188,189].
Whether the analogues of TTX are biosynthesized or formed as metabolites or act as precursors for the synthesis of TTX in affected bearing organisms is yet to be verified.

6. Resistance to TTX in TTX Bearing Organisms

Many researchers have been intrigued as to how TTX bearing organisms are themselves resistant to the toxic effects of TTX. The reason is because in these animals the aromatic amino acid chain in the p-loop region of domain I in the sodium channels is replaced by a non-aromatic amino acid and this prevents the sodium channels in these species from being blocked [103,190,191].
Resistance to TTX can also be related to the presence of a TTX-binding protein present in the shore crab, Hemigrapsus sanguineus [192], in puffer fish, Takifugu niphobles [188] and Fugu pardalis [189,193] and also in five gastropod species: Polinices didyma, Natica lineata, Olivaminiacea, O. Mustelina and O. hirasei [194].

7. Clinical Study

There are four grades of TTX poisoning described by Fukuda and Tani, 1941 [195].
Grade 1: perioral numbness and paresthesia (sensation of tingling, tickling, prickling, pricking, or burning of a person’s skin), with or without gastrointestinal symptoms.
Grade 2: lingual numbness (numbness of face and other areas), early motor paralysis and incoordination, slurred speech with normal reflexes.
Grade 3: generalized flaccid paralysis (reduced muscle tone without other obvious cause), respiratory failure, aphonia (the inability to produce voice due to disruption of the recurrent laryngeal nerve), and fixed/dilated pupils (conscious patient).
Grade 4: severe respiratory failure and hypoxia (inadequacy of oxygen), hypotension (low blood pressure), bradycardia (resting heart rate of under 60 beats per minute), cardiac dysrhythmias (irregular heartbeat) and unconsciousness may occur.
As mentioned previously, the grade of TTX poisoning depends upon the amount of TTX ingested, the time after ingestion of TTX, the hydration state of body and the general health status of the victim prior to intoxication [105].

7.1. Time after Ingestion of TTX

During the Bangladesh outbreak of TTX poisoning in 2008 [19], the onset of symptoms was observed within 30 min of ingestion of puffer fish in 66% of the total number of cases, within 31–60 min in 24% of cases, within 61–90 min in 7% of cases and within 91–120 min in 2% of cases. The poisoning symptoms decreased gradually over 8–28 h after ingestion of the contaminated puffer fish with no residual side effects.
During the TTX poisoning outbreak in Israel between 2005 and 2008, the onset of symptoms was seen within 10–60 min after ingestion. Whereas during the TTX poisoning event in Taiwan in April 2001 [6], the onset of symptoms was typically within 6 h of ingestion, with a delay of up to 20 h in a few cases. The victims became well without any remaining side effects and were discharged 48–72 h after ingestion.
TTX can be found in blood within less than 24 h after its ingestion. But it can be found in urine after 4 days from the time of ingestion [100,196]. Therefore, it is important to collect urine and blood samples from affected patients within 24 h after ingestion for clinical diagnosis.

7.2. Amount of TTX Ingested

The victims of the Bangladesh outbreaks in April 2008 [19] ingested less than 50–200 g of puffer fish. The victims who died had ingested more than 100 g of TTX contaminated puffer fish.
During the Bangladesh outbreak in April 2002 [9], 37 victims were affected by TTX poisoning. There was not any correlation between the quantity of puffer fish ingested and the number of deaths. Surprisingly, 4 deaths were seen among 10 victims who had ingested just 51–100 g of fish but only 3 deaths were seen among those that had ingested larger portions (101–500 g) of the fish.

7.3. Physical Status of Victim

There was no significant correlation between urine TTX levels and blood TTX levels and the severity of the poisoning symptoms found during the Bangladesh outbreak [19]. This is because urine TTX levels depend not only on the amount of TTX ingested but also on the hydration state of the victim. While analysing TTX in urine, it is necessary to do a creatinine adjustment in order to eliminate the effects of the variations in the urinary outputs between patients [135]. Zimmer, 2010 [97] found a strong correlation between blood TTX levels and the poisoning symptoms. This could be due to similar hydration states of the victims.
During the TTX poisoning episode that occurred in Taiwan in April 2001 [106], urine and blood samples of 4 of the victims were collected about 10 h after ingestion and analysed by LC-MS. The blood TTX level of one victim who died was 40.6 nM (12.96 ng/mL) and that of the survivors’ varied between 4.5 and 28.6 nM (1.44–9.13 ng/mL). The urine TTX level of the victim who died was 325 nM (103.71 ng/mL). The urine TTX level of the two survivors was lower (190 nM (60.63 ng/mL) and 47 nM (15 ng/mL)). However a higher level of TTX was found in the urine of one of the survivors (344 nM (109.77 ng/mL)) which was greater than the patient who died (325 nM (103.71 ng/mL)) and both of them had ingested almost the same quantity of fish (100 and 90 g respectively). This suggests that the victim who died might not have excreted the TTX fast enough resulting in higher levels in the blood [40.6 nM (12.96 ng/mL)] which led to death just 4 h after ingestion. All of the survivors recovered well without long term effects. This shows individual variability of patient outcomes with regard to TTX poisoning.

7.4. Health Status of Victim

TTX poisoning was observed in a wide range of age groups in the TTX outbreak in Bangladesh [19]. Therefore, all age groups are susceptible to intoxication by TTX toxin. TTX poisoning in people suffering from diabetic neuropathy (nerve damage), uremia (illness accompanying kidney failure) and Na-K-adenosine-triphosphate deficiency modify the symptoms of TTX poisoning [196]. Diabetic neuropathy is a neurological disorder related to diabetes mellitus. It affects all peripheral nerves including the pain fibres, motor neurons and the autonomic nervous system; therefore it can affect all organs. TTX poisoning in a patient with diabetic neuropathy can lead to severe synergistic effects. During the TTX poisoning incident in Taiwan in April 2001 [6], one of the victims who died from TTX poisoning had diabetic neuropathy. This victim ingested 80 g of puffer fish which was less than the amount ingested by one of the survivors (100 g) without any residual effects.
Na-K-adenosine-triphosphatase (EC 3.6.3.9) [197], also known as the sodium-potassium pump is an anti-porter enzyme present in the plasma membrane of all animal cells. The Na+/K+-ATPase enzyme pumps sodium out of cells, while pumping potassium into cells. TTX poisoning in persons suffering from Na-K-adenosine-triphosphate deficiency can have severe effects.
Uremic syndrome (uremia) [198] is a serious complication of chronic kidney disease and acute renal failure. An excess of urea and waste products accumulate in the body of uremic patients due to kidney dysfunction. This can led to a range of symptoms which are similar to TTX poisoning symptoms including; nausea, vomiting, changes in mental status such as confusion, reduced awareness, agitation, psychosis, seizures and coma, abnormal bleeding such as bleeding spontaneously or profusely following a minor injury, heart problems such as an irregular heartbeat, inflammation and an excess of fluid in the sac that surrounds the heart and increased pressure on the heart. Nakashima et al., 2007 [199] reported mild TTX poisoning in a uremic patient (a 52-year-old woman) from Japan, who had numbness of the lips and weakness in the legs after ingestion of puffer fish. While her symptoms were not severe and she did not require respiratory support, her condition did not improve after 2 days of hospitalization. Haemodialysis (extracorporeal removal of waste products such as creatinine and urea and free water from the blood when the kidneys are in a state of renal failure) was therefore performed on hospital days 3, 4 and 5, and resulted in a marked improvement of symptoms. As a consequence of uremia, TTX accumulated in her body due to its delayed excretion. This suggests that haemodialysis might be helpful for the treatment of TTX poisoning [199].

7.5. Clinical Findings

Clinical manifestations of TTX poisoning had been reported during various outbreaks [6,9,19,105,106]. Kanchanapongkul et al., 2008 studied 280 cases of TTX poisoning caused by ingestion of eggs of the horseshoe crab, Carcinoscorpius rotundicauda, between 1994 and 2006 that were admitted to Chon Buri Hospital in Thailand [172]. Out of 245 medical records; 100 were in stage 1; 74 were in stage 2; 3 were in stage 3 and 68 were in stage 4 of intoxication.
During the Bangladesh outbreak in April 2008 [19], the blood TTX level was >9 ng/mL (28.20 nM) in 7 of the victims who died (5 within 15–30 min and 2 after 4 h of ingestion of puffer fish). Only 3 victims survived, in which the blood TTX level was between 9.3 and 10 ng/mL (29.14–31.34 nM). This suggests that a blood TTX level of 9 ng/mL (28.20 nM) or greater may be considered as potentially lethal for human beings.
In the Bangladesh outbreak [19], routine haematological, biochemical and ECG tests were normal in all of the victims. In the Israel outbreak [105], these tests were also normal in almost all of the victims, however one victim had hypercapnia (too much CO2), one had hypokalemia (low level of potassium in blood which is maintained by the Na+/K+ pump) and two had elevated levels of CPK (creatine phosphokinase) which is a marker enzyme for myocardial infarction (heart attack), rhabdomyolysis (severe muscle breakdown), muscular dystrophy, autoimmune myositides (chronic inflammation of striated muscle) and acute renal failure. In this outbreak [105], one of the victims was likely to have had a seizure (a neurological disorder; often associated with a sudden and involuntary contraction of a group of muscles and loss of consciousness). Previously it had been reported that seizures were correlated with TTX poisoning [38].

7.6. Treatment

There is presently no antidote available for TTX poisoning. Chew et al., 1984 [200] have tried to make use of an anti-cholinesterase drug for treating TTX. During the large TTX incident in Bangladesh [19], 21 victims were given anticholinesterases; neostigmine and atropine but it wasn’t seen to improve their condition. This is because anticholinesterases reverse the blocking action at neuromuscular junction at the motor end plates only. Whereas TTX blocks sodium channels of motor neurons and muscle membranes.
One of the severely poisoned victims of the TTX outbreak in Israel [105], had been administered 0.4 mg of naloxone intravenously which showed an equivocal response. Naloxone has a high affinity for μ-opioid receptors in the central nervous system and is used for treating depression of the central nervous system and respiratory system.
Currently, the only treatment for TTX poisoning is to provide the victim with respiratory support until the TTX is excreted completely. Endotracheal intubation can be provided to facilitate ventilation of the lungs. Mechanical ventilation may also be provided. During the TTX poisoning outbreak in Israel [105], patients were given respiratory support and recovered within 4 days. During the TTX poisoning event in Thailand [172], victims were treated with endotracheal intubation and mechanical ventilation. Out of 245 victims, 239 patients (97.5%) completely recovered, 5 patients (2%) died, and 1 patient (0.4%) suffered anoxic brain damage.
In the case of early stage TTX poisoning victims are given activated charcoal in order to help the adsorption of TTX to prevent its absorption through the stomach [201]. Gastric lavage (the passage of a tube via the mouth or nose down into the stomach followed by sequential administration and removal of small volumes of liquid) can be performed in TTX poisoning in order to reduce its severity. This procedure should be performed within 60 min after ingestion of TTX [202]. In the Bangladesh outbreak in April 2002 [9], 37 patients were given gastric lavage and neostigmine treatment along with respiratory support. But there are some risks associated with gastric lavage treatment such as laryngospasm (involuntary muscular contraction of the laryngeal cords), hypoxia (inadequacy of oxygen), bradycardia (a resting heart rate of under 60 beats per minute), epistaxis (nosebleed), hyponatremia (reduced levels of sodium in the blood), hypochloremia (reduced levels of chloride ions in blood), water intoxication or mechanical injury to the stomach.
Intravenous fluids are also given in order to maintain fluid-electrolyte balance in the body during TTX poisoning. An antiemetic is given which is effective against vomiting and nausea. As mentioned in paragraph 7.4, haemodialysis might also be useful for the treatment of TTX poisoning [199].
Antibodies against TTX have been used successfully in vivo [203,204]. Xu et al., 2005 synthesized antibody against TTX [130]. This antibody was able to neutralise the toxic effect of TTX both in vitro and in vivo. A monoclonal antibody for TTX (anti-TTX) is available commercially from Hawaii Biotech, Inc., Aiea, HI, USA [121,136]. However, studies on the efficacy of this monoclonal antibody in vivo have not been published [121], but this may herald the advent of a new type of approach to the treatment of TTX poisoning in the future.

7.7. Application of TTX in the Medical Field

Some researchers are trying to make use of the analgesic activity of TTX to treat various types of pains such as severe cancer pain [205,206,207]. A low dose of TTX has also been shown to help in reducing cue-induced increases in heroin craving and associated anxiety [208].

8. The Distribution of TTX and Its Analogues

8.1. Geographic Distribution of TTX

8.1.1. Geographic Distribution of TTX in Freshwater and Marine Organisms

TTX poisoning is very common in Japan, Taiwan, Bangladesh and Southeast Asia (Table 1). Most commonly it is associated with the consumption of puffer fish and sometimes by the ingestion of gastropods or crabs. In the last decade, TTX was found in puffer fish [29,48,131] and gastropod [44,45,48] collected from European countries as well. A number of researchers have formulated the theory of “Lessepsian migration” for explaining the new occurrence of TTX in European regions. In 1869, the opening of Suez canal caused migration of many Red Sea species through the new waterway which settled in the Eastern Mediterranean e.g., occurrence of Lagocephalus sceleratus in the Mediterranean Sea. Hence, this phenomenon is called “Lessepsian migration” [105,131,209]. The occurrence of several migrant marine species have been reported from Mediterranean Sea such as Lagocephalus sceleratus, Mobulamobular, Tylosuruschoram, Scarusghobban and Tetrapturus belone [210,211,212,213,214,215,216]. Ballast water can also cause the transfer of TTX containing organisms from Asian waters to European waters. Over the last 20 years, spreading of marine mucilage in the Mediterranean Sea was observed due to sea surface warming [99]. This helps the survival of migrated species in Mediterranean Sea.
Some cases of TTX poisoning caused by the consumption of puffer fishes have been observed in the US also [2,108,110]. But those species of fish were imported from either Japan or from other places. TTX had been found in puffer fishes, Sphoeroides annulatus, S. lobatus, S. lispus, Arothron meleagris and Canthigaster punctatissima collected from the coast of the Baja in the California Peninsula, Mexico [168]. Mexico is the second largest puffer fish exporter in the world.
Occasionally natural calamities can also cause small changes in the marine ecology of a region and thus the occurrence of new and/or displaced species. Chulanetra et al., 2011 [27] state that the ecology of marine organisms including puffer fish might have been affected by the Tsunami in the Andaman Seas which originated from the earthquake in the Indian Oceans in 2004. They studied the toxicity of 155 puffer fishes caught off the Gulf of Siam and from the Andaman Sea near Thailand, during April to July 2010. Among 125 puffers from the Gulf of Siam, 18 were Lagocephalus lunaris and 107 were L. spadiceus which were found previously in 2000–2001 in the same region. They also reported the presence of TTX in L. spadiceus for first time. In 1992–1993, nine marine puffer fish species were caught in the Andaman Seas, Arothron immaculatus, A. stellatus, Chelonodon patoca, Diodon hystrix, Lagocephalus lunaris, L. inermis, L. spadiceus, L. sceleratus, and Xenopterus naritus. Out of these, five puffers, A. immaculatus, C. patoca, L. lunari, L. sceleratus and X. Naritus were found to be positive for TTX [27]. In 2011, thirty puffers were collected from the Andaman Sea: 28 Tetraodon nigroviridis and two juvenile Arothron reticularis; the two new species totally replaced the nine species found previously in the same region during the 1992–1993 study and some of these were found positive for TTX [27].

8.1.2. Geographic Distribution of TTX in Terrestrial Animals

TTX was found in newts from the USA, Japan, Germany and Italy; Notophthalmus viridescens from Canada and USA [37,51,53,54,55,57,58], Notophthalmus viridescens and Taricha from North America [36], Cynops ensicauda popei from Okinawa, Japan [59], Cynops pyrrhogaster from Japan [52], Triturus spp. from Germany [35] and Triturus alpestris from Italy [32].
TTX was also found in frogs belonging to the Brachycephalidae family from the Brazilian Atlantic rain forest [33,72], from the Central American frog Atelopus chiriquiensis [30] and in the Rhacophoridid frog, Polypedates sp. from Bangladesh [71].

8.2. Organism Specific Distribution of TTX and Its Analogues

The occurrence of 26 natural analogues of TTX had been reported to-date (Figure 1). The name tetrodotoxin is based on the name of the order of animals called tetradontiformes first suggested by Dr. Yoshizumi Tahara in 1909. He isolated TTX from the ovaries of globefish, Fugu [20]. Up to 1964, TTX was believed to be present only in puffer fish. In 1964 Mosher [217] found TTX in the California newt, Taricha torosa. Subsequently, it was also found in frogs, shell fish, star fish, ribbon worms, sea slugs and bacteria (Table 3).

8.2.1. Distribution of TTX and Its Analogues in Puffer Fish

TTX is found in marine, fresh water and brackish water organisms [93]. The presence of TTX in puffer fish is well documented. Table 3 gives details of puffer fish species containing TTX and its analogues. Puffer fish of the Tetradontidae family are toxic while puffer fish from the Diodontodae and Ostracitidae families are usually non-toxic [93]. Lagocephalus wheeleri, L. gloveri and Takifugu xanthipterus are considered as non-toxic species of puffer fish [93,218]. Nakashima et al., 2007 [199] reported mild TTX poisoning caused by Lagecephalus wheeleri in Japan for the first time. Simon et al., 2009 [219] also found TTX in Lagecephalus wheeleri from Malaysian waters identified by LC (liquid chromatography) but it was not lethal to mouse in the mouse bioassay. Nagashima et al., 2001 [218] have shown that non-toxic Takifugu xanthipterus contains less toxic TTX derivatives. This indicates that TTX might be converted into less toxic derivatives in these species. T. nigroviridis was more toxic than Lagocephalus lunaris, L. Spadiceus and Arothron reticularis collected from the Andaman seas [27].
Female puffer fish are more toxic than male puffer fish as they accumulate TTX in the ovaries and eggs during the spawning period [46]. Jang et al., 2006 [24] studied distribution of TTX analogues in Fugu pardalis. 5,6,11-trideoxyTTX (especially in ovaries) was the major and 5-deoxyTTX and 11-deoxyTTX were minor TTX analogues in all tissues. Whereas 4,9-anhydroTTX was the major analogue in liver, 4-S-CysteinylTTX was detected in liver, spleen, gall, and intestine in 1–6 mole percentage of the total of all the TTX analogues.
Vázquez et al., 2000 [168] found that 4,9-anhydroTTX was the major and 4-epiTTX was minor analogue of TTX in puffer fishes, Arothron meleagris (black phase), A. meleagris (golden phase), Sphoeroides annulatus and S. lispus. 4,9-anhydroTTX was not found in both S. lobatus and Canthigaster punctatissima whereas, 4-epiTTX was found in very low concentration in Canthigaster punctatissima. In general, the level of TTX was higher than the level of 4,9-anhydroTTX in liver. While the level of 4,9-anhydroTTX was higher than the level of TTX in mucus. Diener et al., 2007 [138] found that TTX was the major analogue in liver, muscle and skin whereas trideoxyTTX was the major analogue in ovaries of Takifugu oblongus. Jang et al., 2010 [26] found that TTX and 5,6,11-trideoxyTTX were the major TTX analogues whereas 4-epiTTX 4,9-anhydroTTX, 5-deoxyTTX and 11-deoxyTTX were minor TTX analogues in Fugu niphobles, Tetraodon nigroviridis and Tetradon biocellatus. 6,11-dideoxyTTX was the major analogue in almost all tissues of F. niphobles, but it was the minor analogue in Tetraodon nigroviridis and Tetradon biocellatus. Kudo et al., 2012 [59] found that the levels of TTX, 5,6,11-trideoxyTTX and anhydro-5,6,11-trideoxyTTX were highest among all the analogues of TTX. Whereas, the level of 4,9-anhydroTTX was highest among all the remaining minor analogues of TTX; 4-epiTTX, 5-deoxyTTX, 11-deoxyTTX, 6,11-dideoxyTTX and 11-norTTX-6(S)-ol in Fugu poecilonotus. Rodríguez et al., 2012 [29] found that 5,6,11-trideoxyTTX was the major TTX analogues, followed by 11-deoxyTTX, 11-norTTX-6(S)-ol, and TTX. While 4-epiTTX, 4,9-anhydroTTX, 5-deoxyTTX and 11-norTTX-6(R)-ol were minor analogues of TTX in Lagocephalus sceleratus.

8.2.2. Distribution of TTX and Its Analogues in Gastropod

TTX and some of its analogues were found in gastropod species: Gibbula umbilicalis- monodeoxy (TTX (0.063 µg/g)); Monodonta lineata (TTX (0.090 µg/g) and 4-epiTTX (0.021 µg/g)) and C. lampas (5,6,11-trideoxyTTX (0.006 µg/g)) [47]; C. lampas (TTX (315 µg/g) and 5,6,11-trideoxyTTX (1004 µg/g)) [45]; N. nitidus (TTX (1350 µg/g)) [25]; N. semiplicatus (TTX (26.1 µg/g) and 4-epiTTX (3.37 µg/g)) [86] and N. papillosu (TTX (42–60 µg/g)) [12]. TTX and its analogues trideoxyTTX, 4-epiTTX, anhydroTTX and oxoTTX were detected in the nassariid species [28]. Usually, the levels of TTX in gastropod are lower than those in puffer fishes. But gastropods are also capable of inducing TTX poisoning [4,5,7,8,10,12,42,45,46]. In general TTX and 5,6,11-trideoxyTTX are the major while 4-epiTTX is the minor analogue of TTX found in gastropods.

8.2.3. Distribution of TTX in Sea Slug, Star Fish, Blue-Ringed Octopus, Ribbon Worm and Bacteria

TTX had been occasionally found in the sea slug, star fish and ribbon worm. TTX was detected in sea slugs, Pleurobranchaea maculata which caused poisoning in dogs on the beaches of Hauraki Gulf, Auckland, New Zealand [73]. The presence of TTX in sea slugs, Pleurobranchaea maculata have also been reported by Wood et al., 2012 [74]. Along with TTX, low levels of 11-norTTX have been found in Pleurobranchaea maculata by McNabb et al., 2010 [73].
TTX has been found in a starfish species, Astropecten scoparius [75], a ribbon worm, Cephalothrix linearis (Nemertean) [22,79] and blue-ringed octopuses, Hapalochlaena fasciata and Hapalochlaena lunulata [76] (Table 3). TTX, 4epi-TTX and anhydroTTX were found in ribbon worm, Cephalothrix sp. present on the shells of cultured oysters [22]. No other analogue of TTX has been reported in either starfish, or blue-ringed octopus todate.
TTX and anhydroTTX were found in bacteria, Shewanella woodyi and Roseobacter sp. isolated from the copepod Pseudocaligus fugu parasitic on the panther puffer Takifugu pardalis [85]. TTX, 4-epi-TTX, and anhydroTTX were found in Vibrio Strain, LM-1, from the puffer fish Fugu vermicularis radiatus [91].

8.2.4. Distribution of TTX and Its Analogues in Terrestrial Animals

As previously stated, TTX can be found in several species of newts: Notophthalmus [32,36,37,50,53,57], Cynops [31,32,52,59] and Triturus spp. [35], crabs: Demania cultripes, Demania toxica, D. reynaudi, Lophozozymus incisus, L. Pictor and A. germaini [62,139,170] and in frogs and toads: Atelopus [70,143] and Brachycephalus [33,34,72].
Along with TTX, other related analogues were also found in newt and salamander: TTX [32,51,54], 6-epiTTX [32,36], 11-oxoTTX [36], 4-epiTTX [51], 4,9-anhydroTTX [51] and 11-deoxyTTX [32] in Taricha spp. Recently four new analogues, 8-epi-5,6,11-trideoxyTTX (MW 271.2710), 4,9-anhydro-8-epi-5,6,11-trideoxyTTX (MW 253.2557), 1-hydroxy-8-epi-5,6,11-trideoxyTTX (MW 287.2704) and 1-hydroxy-4,4a-anhydro-8-epi-5,6,11-trideoxyTTX (MW 269.2551) have been isolated from newt which are not present in puffer fish [59].
Pires Jr. et al., 2002 [33] found TTX, 4-epiTTX, 4, 9-anhydroTTX, 11-norTTX-(S)-ol and tetrodonic acid in the frog, Brachycephalus ephippium. Pires Jr. et al., 2003 [34] identified 11-oxoTTX also from the same species of frog. Pires Jr. et al., 2005 [72] have studied the toxicity of three species of frogs: Brachycephalus ephippium, B. nodoterga and B. Pernix belonging to the Anuran family, Brachycephalidae. Toxicity was highest in the skin followed by the liver and ovary in Brachycephalus ephippium and B. prenix. 11-oxoTTX, TTX, 4-epiTTX, 4,9-anhydroTTX, 5-deoxyTTX and tetrodonic acid were found in Brachycephalus ephippium while TTX, 4,9-anhydroTTX, 5-deoxyTTX and tetrodonic acid were found in B. Prenix by LC-FLD (liquid chromatography-fluorescent detection). Traces of 11-oxoTTX, TTX, 4-epiTTX, 4,9-anhydroTTX, 5-deoxyTTX and tetrodonic acid were found in B. nodoterga using LC-FLD but B. nodoterga extract was non-toxic by mouse bioassay. Also, two unknown compounds having mass spectral signals at m/z 330 and 348 which may be attributed to TTX analogues but could not be unequivocally identified. The 11-oxo TTX analogue is commonly found in frogs but rarely seen in puffer fish and newts.
TTX and anhydroTTX were found in xanthid crab, Xanthias lividus [67]. Along with TTX small amounts of anhydroTTX were also found in crab, D. cultripes [139]. TTX was the major while 4-epiTTX and 4,9-anhydroTTX were minor analogues of TTX in xanthid crab, Demania cultripes [170]. 11-oxotetrodotoxin and 11-nortetrodotoxin-6(R)-ol have been reported in a xanthid crab, Atergatis floridus [62].
6-epi TTX is more commonly found in terrestrial organisms than in marine or in fresh water species. Table 3 gives details of the occurrence of TTX and its analogues in a variety of organisms.

8.3. Tissue Specific Distribution of TTX and Its Analogues in All Organisms

Puffer fish: TTX is found in all tissues of puffer fish. The levels of TTX vary among species of puffer fish. A study of the tissue distribution of analogues of TTX in Fugu Pardalis was compiled by Jang and Yotsu-Yamashita in 2006 [24]. They found that the liver and ovary are more highly toxic than the muscle and testis in wild puffer fish. 4-Cys TTX was found in the liver, spleen, gall bladder and intestine of Fugu Pardalis but not in the ovary [24].
In marine puffer fish, the liver tissue is toxic throughout the year except during the spawning season but the ovaries become more toxic perhaps to protect the organism from predators. In puffer fish that inhabit brackish water and freshwater regions toxicity is higher in the skin than in marine species [93].
The average level of 4-epiTTX in puffer fish is ca. 10% (mol/mol) of TTX [128] and 5,6,11-trideoxy TTX was the major analogue in all tissues of the puffer fish, Fugu pardalis [125]. The levels of TTX analogues were highest in the ovary of puffer fish Takifugu oblongus than in any of the other tissue compartments. Among all the analogues, the levels of 5,6,11-trideoxyTTX was the highest and was located in the ovaries [138].
Gastropod: TTX is mainly found in the muscles of gastropods. Some amounts of TTX are also found in the digestive glands. TTX and its analogues trideoxyTTX, 4-epiTTX, anhydroTTX and oxoTTX were detected in the gastro pod nassariid [28]. TrideoxyTTX was the major toxin in all the samples [28]. The toxicity of TTX was higher in the muscle tissue than in the digestive gland in N. lineata [184]. The toxicity of edible portions of Polinices didyma and of Natica lineate was higher than that in the digestive gland [8].
Newt: TTX is found mainly in the skin of newt. In newt (Taricha granulosa), TTX levels were high in the skin of adult newt and in the yolk of embryo. TTX levels decrease during the development of larvae and larvae are almost non-toxic [58].
Crab: Less information is available regarding tissue specific distribution of TTX and its analogues in crab. TTX has been found in xanthid crabs, Demania cultripes, Demania toxica, D. reynaudi, Lophozozymus incises, L. Pictor and A. germaini in southern Taiwan. Toxicity was higher in the viscera (4.0–11.9 MU/g) than in the appendages and cephalothorax in all of the species tested. Toxicity of the appendages was 1.6–12.3 MU/g while toxicity of cephalothorax was 1.5–3.6 MU/g among all the species [139].
Frog: Skin is the main toxic tissue in frog. Pires Jr. et al., 2005 [72] have studied the toxicity of three species of frogs; Brachycephalus ephippium, B. nodoterga and B. pernix belonging to the Anuran family, Brachycephalidae. Toxicity was highest in the skin followed by the liver and ovaries in Brachycephalus ephippium and B. prenix which was studied by mouse bioassay. They have not given the toxin profile of individual tissues. Chiriquitoxin (CqTX) was found in the skin and in the eggs of Atelopus chiriquiensis [30,70].
Other organisms: TTX was found in the skin of the gobies [220] and in the head of arrow worms [221] and in the proboscis of the ribbon worm [79]. Blue-ringed octopuses contain TTX in the posterior salivary gland, the skin and eggs. Williams and Caldwell, 2009 [76] studied 14 tissue types from blue-ringed octopuses. In their study TTX was found in the posterior salivary gland (PSG), arm, mantle, anterior salivary gland, digestive gland, testes contents, brachial heart, nephridia, gill and oviducal gland of Hapalochlaena fasciata. But in H. lunulata TTX was found only in the PSG, mantle and ink.

8.4. TTX Co-occurrence with Other Marine Toxins

Occurrence of either minor amounts of paralytic shellfish poisons (PSP) along with TTX, or major amounts of PSP along with traces of TTX have been reported in many species of puffer fish, crabs and gastropods (N. Clathrata, Polinices didyma and N. Lineata) [8]. Sometimes species that are susceptible to TTX intoxication have been found to be free of TTX (or have TTX only at very low levels) while they have been found to have other toxins such as PSP present at the time of testing. Hydroxysaxitoxin has been found in the xanthid crab, Demania cultripes from the Philippines [170]. Saxitoxin, decarbamoyl saxitoxin have been confirmed in Cambodian freshwater puffer fish, Tetraodon turgidus (which is not resistant to TTX) [222]. The presence of GTX-2 and GTX-3 has been noted in Colomesusasellus, an Amazonian (Brazil) freshwater puffer fish [223]. Saxitoxin and decarbamoyl saxitoxin have been found in Fugu pardalis [24]. Saxitoxin and decarbamoyl saxitoxin have been identified in the marine puffer Arothron firmamentum [167]. GTX-1,2,3 and 4 have been found in the Taiwanese crab, Xanthias lividus [67]. The puffers Sphoeroide snephelus, S. testudineus, and S. Spengleri have tested positive for saxitoxin from the Indian River Lagoon, Florida [224]. Gonyautoxin 2, gonyautoxin 3 and saxitoxin (STX) and neoSTX, decarbamoylSTX and STX have been found in the xanthid crab Zosimus aeneus [66]. Saxitoxin, decarbamoyl saxitoxin, gonyautoxins 2 and 3, decarbamoyl gonyautoxins 2 and 3 have been found in the freshwater puffer fish, Tetraodon cutcutia and Chelonodon patoca from Bangladesh [225]. Gonyautoxin 1-4 in the xanthid crab, Atergatis floridus and gonyautoxin 2-4 and neosaxitoxin in the xanthid crab, Demania reynaudi from Taiwan [65], saxitoxin, neosaxitoxin and decarbamoyl saxitoxin in the freshwater puffers, Tetraodon leiurus and Tetraodon suvatii from Thailand [226], gonyautoxin-3; GTX3, GTX2, and saxitoxin in the gastropod Rapana venosa from Japan [227], saxitoxin in the freshwater puffer, Tetraodon fangi from Thailand [228], and saxitoxin in the southern (Sphoeroide snephelus), checkered (Sphoeroide stestudineus), and band tail (Sphoeroide spengleri) puffer fish from the US [229] were found. TTX was found to co-occur in small quantities with PSP in A. Germaini in northern Taiwan. TTX and GTX were also found to co-occur in D. reynaudi and L. pictor in northern Taiwan [139]. TTX was found in samples together with palytoxin in Demania cultripes, D. reynaudi and L. pictor of Philippine and in L. pictor from Singapore [139].
Both, TTX and PSPs (Saxitoxin, neosaxitoxin, decarbamoyl saxitoxin, hydroxysaxitoxin, gonyautoxin 1, 2, 3 and 4) are neurotoxins which block the sodium channels. The structures of TTX (Figure 1) and STX (Figure 5) are similar; they bind to a common site, which is present at the external mouth of the sodium channels [230]. Symptoms of paralytic shellfish poisoning are similar to TTX poisoning [201]. So there is a risk of misdiagnosis in such poisoning events. Several treatments for TTX poisoning have been discussed earlier in the text (Section 7.6). Interestingly, 4-aminopyridine was observed to reverse the effect of saxitoxin and tetrodotoxin in mice without any side effects such as seizure or convulsions [231,232]. The optimal 4-AP dose was determined as 2 mg/kg (im) [232].
Figure 5. Structures of paralytic shellfish poisons (PSP) toxins.
Figure 5. Structures of paralytic shellfish poisons (PSP) toxins.
Toxins 06 00693 g005

9. Chemical Stability and Toxicity of Analogues

TTX is heat stable and water soluble [233]. It is an aminoperhydroquinazolone. 4-epiTTX and 4,9-anhydroTTX are in chemical equilibrium with TTX while deoxy analogues (5-deoxy TTX, 11-deoxy TTX, 6,11-dideoxy TTX and 5,6,11-trideoxyTTX), 11-oxo TTX and 4-S-cysteinyl TTX are not in chemical equilibrium with TTX [26,89,166].
The toxicity of analogue is based on the number and position of hydroxyl groups present in the structure. Yotsu-Yamashita et al., 1999 [112] have studied the effects of specific modifications of a number of hydroxyls of TTX on its affinity to rat brain membrane. The results showed that hydroxyls at C-6 and C-11 play an important role in the binding of these toxins to the sodium channels, acting as hydrogen bond donors. In an earlier study, Yang and Kao showed that hydroxyls at C-4, C-6, C-9, C-10 and C-11 also play role in binding to the sodium channel [70].
11-OxoTTX is 4 to 5 times more toxic than TTX [34]. The additional OH group of 11-oxoTTX probably binds to the sodium channels with hydrogen bonding more efficiently than TTX. 11-oxo TTX is rare in marine animals and it is found predominantly in frogs.
5-Deoxy TTX, trideoxy TTX, 4-Cys TTX and anhydro TTX have negligible toxicity. The toxicity of 5,6,11-trideoxyTTX is less than that of TTX as it has less hydroxyl groups compared with that of TTX. Fewer hydroxyl groups serve to reduce the binding affinity of 5, 6, 11-trideoxyTTX to the sodium channels. Jang and Yotsu-Yamashita, 2007 [125] have found that the toxicity of 6,11-dideoxyTTX was higher than a synthetic analogue 8,11-dideoxyTTX.
Jang and Yotsu-Yamashita, 2007 [24] make the assumption that 4-CysTTX might be the metabolite of 4,9-anhydroTTX formed by the reduction of glutathione as they have found high levels of 4,9-anhydroTTX and 4-CysTTX in liver, spleen, gall and intestine but not in the ovaries of selected species.
Kudo et al., 2012 [59] hypothesized that TTX is the oxidation product of 5,6,11-trideoxyTTX in the TTX-producing marine organisms.
Kono et al., 2008 [166] and Wang et al., 2011 [134] state that TTX is metabolised in the puffer fish, Fugu niphobles and thus produces different analogues; most abundantly 4,9-anhydroTTX. In puffer fish TTX accumulates in the liver and then slowly transfers to the skin.

10. Miscellaneous Studies

Saoudi et al., 2007 [15] showed that in the cooked flesh together with the water of cooking, the toxic effect was more pronounced than in the raw flesh. Shiu et al., 2003 found that the level of TTX in cooked gastropod, Polinices didyma was in the range of 30–261 MU/specimen [8], while Taylor et al., 2011 found TTX levels in the range of 374–655 µg/100 g in the cooked flesh and in the soup broth [136]. This indicates that cooking does not remove or degrade TTX. Anraku et al., 2013 [234] have found that the traditional salting and fermentation process does reduce TTX levels in the ovaries of puffer fish, Takifugu stictonotus.

11. Historical Perspective on Analytical Methods Used for TTX and Its Analogues

11.1. Bioassays

Receptor binding assay [235]; immunological methods (e.g., ELISA) [23,52,88,121,134,236,237,238] and mouse bioassay [27,28,81,92,131,223,239] have all been used for TTX analysis. The mouse bioassay is the method that is most frequently applied. All of the above methods can be used successfully to identify TTX but not all of its analogues. Researchers tried to improve the accuracy and repetition of bioassays. Recently Stokes et al., 2012 [240] have developed a competitive inhibition enzymatic immunoassay method which has high accuracy and repeatability at concentration range of 10–100 ng/mL. However, this method doesn’t identify any of the other analogues of TTX and also it involves the use of expensive antibodies (primary and secondary). There are also ethical issues regarding the use of live animal bioassays (the mouse bioassay) when more accurate and precise analytical methods that can both qualitatively and quantitatively profile TTX and its analogues are widely available.

11.2. Chemical Assays

Surface plasma resonance [136], electrophysiological assays [97,98,191,232]; IR [96]; NMR [53,59,96,125,173,241,242]; GC-MS [7,8,85,137,139,169,170,171]; LC-FLD and LC-MS (Table 4, Table 5, Table 6, Table 7, Table 8 and Table 9) have been developed and used for the determination of TTX by many researchers. In addition NMR, GC-MS, LC-FLD and LC-MS provide the benefit of finding not only TTX but related analogues along with co-occurring toxins in samples thus allowing a more comprehensive risk assessment of produce intended for human consumption.
Yotsu-Yamashita et al., 1999 have quantified 6-epiTTX, 11-deoxyTTX and 5,6,11-trideoxyTTX by 1H-NMR using TTX standard [112]. The difficulty with NMR is that in real samples intense interference from matrix components can compromise the quality of the spectra.
Due to huge variations in fluorescence intensities of different members of the TTX family as well as interfering background signals from the matrix, LC-FLD is not a good choice for the routine analysis of real samples. The fluorescence intensities of 6-epiTTX and 11-norTTX-6(R)-ol are 20-fold and 10-fold higher respectively than that of TTX while the fluorescence intensities of 5-deoxyTTX and 11-deoxyTTX are 1/20 and 1/100 lower than that of TTX [72,149].
GC-MS has been used to screen for the presence of TTX. However because TTX is non-volatile, it needs to be converted into its volatile derivative before analysis by GC-MS. This is a disadvantage for the method as derivatisation requires a large amount of sample, and the method suffers from poor reproducibility and is also time consuming.
Therefore LC-MS and especially LC-MS/MS are generally regarded as the best choice for the determination of TTX and related compounds (Table 4, Table 5, Table 6, Table 7, Table 8 and Table 9).

11.3. Historical Overview of LC-MS/MS Methods for TTX and Its Analogues

Table 4, Table 5, Table 6, Table 7, Table 8 and Table 9 provide a brief summary of the LC-MS methods that have been used for the analysis of TTX and its analogues in a variety of sample types: puffer fish (Table 4), trumpet shell/ gastropod, sea slug and octopus (Table 5), newt (Table 6), crab and frog (Table 7), bacteria (Table 8) and Human blood/ urine (Table 9).

11.3.1. Extraction and Clean Up Methodologies

Table 10 summarises the extraction processes and percentage (%) recoveries of TTX from different matrices. The % recovery varies from 80% to 90% for most of the extraction protocols when applied to real samples. However, the recovery of TTX was only 50% for newt samples [35].
Table 4. LC-MS methods for TTX and its analogues from puffer fish.
Table 4. LC-MS methods for TTX and its analogues from puffer fish.
SpeciesExtractionColumnMobile phaseMethodAnalyte *LOD and LOQLinear RangeReference
Lagocephalus sceleratus (Gmelin, 1789)
(Liver, GI-tract, gonad (ovary/testis), muscle and skin)
0.1% AAZorbax 300SB-C3
Sunfire C18
XBridge™ Amide
Isocratic: 1% ACN + 10mM TMA + 10 mM AF, pH 4 (For Zorbax 300SB-C3)
A: 1% ACN + 20 mM AHB + 20 mM Am-OH + 10 mM AF, pH 4 and B: 5% ACN + 20 mM AHB + 20 mM Am-OH + 10 mM AF, pH 4 (For Sunfire C18)
A: 10 mM AF + 10 mM FA in H2O
B: 5 mM AF + 2 mM FA in ACN:H2O, 95:5 (For XBridge™ Amide)
LC-MS/MS and CID-MS/MS6, 7, 11, 12, 14, 17, 18, 22LOD:16 ng/mL at S/N > 3
LOQ:63 ng/mL at S/N > 10
62.5–2000 ng/mL[29]
Lagocephalus sceleratus (Muscle)ASE and SE (0.03 M AA)Acquity UPLC BEH HILICA: 5% ACN
B: 95% ACN + 1% AA pH 3.5
LC-M/MS6For Solvent Std LOD: 0.074 ng/mL
LOQ: 0.123 ng/mL
For Matrix-matched Std
LOD: 7.3 µg/kg and LOQ: 24.5 µg/kg at S/N = 3 and 10
5–500 ng/mL (Solvent Std)
50–3000 µg/kg (Matrix-matched Std)
[48]
Potka or Tepa fish (Cooked fishAnd blood and urine of victim)1% AA + 80% MeOHC30 UG-5A: 30 mM AHB, pH 5 in H2O
B: 10 mM AF, pH 5 in 1% ACN
LC-FLD6, 7, 14NRNR[19]
Lagocephalus lunaris, L. spadiceus, Tetradon nigroviridis and Arothron reticularis (Reproductive tissue, digestive tissue, liver, muscle and skin)0.1% AA, ethyl acetate Defat *, CharAd ZIC-HILICA: 10 mM AF + 10 mM FA in H2O
B: 5 mM AF + 2 mM FA in 80% ACN
LC-MS/MS6, 11/12, 16NRNR[27]
Puffer fish (ovary)0.05 M AA, ODS-SPE, Ultra filtration (0.22 µ)Atlantis HILIC Silia10 mM AF, pH 3.5 + ACN (22:78, v/v)LC-MS (SIR)
LC-MS/MS (CID)
6, 7, 9, 11, 14, 17/ 18, 20, 22SIR mode
LOD: 0.1 ng/mL at S/N = 3
LOQ: 0.25 ng/mL at S/N = 10
0.25–100 ng/mL[127]
Takifugu rubripes and Takifugu niphobles (Muscle, skin, liver, gonad)1% AAPuresil C1830 mM HFB + 1 mM Am-acetate, pH 5.0LC-MS6, 7, 14NRNR [167]
Fugu niphobles (Ovary/testis, liver, intestine, dorsal skin and dorsal muscle)
Tetraodon nigroviridis and Tetraodon biocellatus (Whole body)
0.05 M AA, C18-SPE, CHCl3 Defat *, CharAd TSKgel Amide-8016 mM AF, pH 5.5 in ACN (3:7, v/v)LC-MS/MS6, 7, 11, 12, 14, 20, 22NRNR[26]
Lagocephalus sp.
(Cooked fish)
1% AA, CHCl3 Defat *TSK-GEL Amide-805 mM AF + 26.5 mM FA in ACN:H2O, 70:30LC-MS6NRNR[108]
Tetraodon turgidus and Tetraodon sp. (Skin, muscle, liver, intestine, gonad; ovary/testis)NMRP-181 mM TBA-PO4, pH 5.8HPLC-FLDPSP toxins (STX, neoSTX, GTX1–4, dcSTX, dcGTX2 and 3)NRNR[222,243]
Fugu poecilonotus (Liver)
Fugu niphobles (Whole body)
Sephadex G-10 Gel filtration (For F. poecilonotus)
0.1% AA, 50% CharAd (For Fugu niphobles)
For F. poecilonotus:
ODS-5; LC-FLD
For Fugu niphobles:
HILIC; LC-MS
5 mM AHB + 50 mM Am-acetate, pH 5 in 3% ACN; LC-FLD (For F. poecilonotus)
5 mM AHB + 50 mM Am-acetate, pH 5 in 3% ACN; LC-FLD (For Fugu niphobles)
LC-FLD
LC-FLD and LC-MS
1, 4, 6, 7, 14 (on ODS-5) and 11, 12, 20, 22 (on HILIC)NRNR[89,166]
Fugu niphobles (Liver, intestine, gonad, bone, muscle, skin, other organs; viscera)0.1% AAODS-520 mM AHB, pH 5 + 10 mM Am-acetate, pH 5 in 3% MeCNLC-FLD6, 7, 14NRNR[89]
Takifugu oblongus (Liver, gonad; ovary/testis, muscle, skin, other organs; viscera)0.1% AASeQuant ZIC- HILICA: 10 mM AF + 10 mM FA in water
B: 5 mM AF + 2 mM FA in 80:20 ACN:H2O
LC-MS/MS6, 7, 12, 16, 22LOD:0.09 ng (TTX), 0.14 ng(AnhydroTTX), 0.20 ng (11-deoxy TTX)0.25–10 ng (TTX)
0.25-5.8 ng (AnhydroTTX)
0.20-5 ng (11-deoxy TTX)
[138]
Fugu pardalis (Ovary)0.05 M AA, EtOAc Defat *, CharAd , Bio-Gel P2 and Hitachi gel 3011C filtrationTSK gel Amide-8016 mM AF, pH 5.5 in ACN (3:7, v/v)LC-MS6, 7, 11, 12, 14, 17, 20, 22NRNR[24,125]
Fugu pardalis (Ovary, testis, liver, spleen, gall, skin, intestine, kidney and muscle)0.1% AA, Cosmosil 75 C18-OPN resinC30 UG-5 (LC-FLD) TSK-GEL Amide-80 (LC/MS)30 mM AHB + 10 mM AF in 1% ACN, pH 5 (C30 UG-5)
16 mM AF, pH 5.5 in ACN (3:7, v/v) (TSK-GEL Amide-80)
LC-FLD
LC-MS
1, 6, 11, 12, 14, 22, LOD 0.07 pmole (LC-MS)NR[24,149,175]
Fugu poecilonotus and F. pardalis (Ovary)0.05 M AA, CharAd C30 UG-5 (LC-MS/MS and LC-FLD)20 mM AHB + 10 mM AF in 1% ACN, pH 4 (LC-MS/MS)
30 mM AHB + 10 mM AF in 1% ACN, pH 5 (LC-LFD)
LC-MS/MS
LC-LFD
6, 7, 10, 11, 14, 17, 18, 22LOD 0.7 pmol at S/N 2 50–1000 pmol [149]
Takifugu xanthopterus (Liver)0.05 M Tris-Ac, pH 8.2; Sephacryl S-400 column filtration, 0.03 M AA, DCM Defat *, Bio-Gel P-2 filtrationYMC AM OSD[218]LC-FLD6, 7, 9, 11NRNR[218]
Note: (Analyte*) Refer to Table 3 for the name of the TTX analytes.
Table 5. LC-MS methods for TTX and its analogues from trumpet shell, gastropods and octopus.
Table 5. LC-MS methods for TTX and its analogues from trumpet shell, gastropods and octopus.
SpeciesExtractionColumnMobile phaseMSAnalyte *LOD and LOQLinear RangeReference
Trumpet shell
Charonia lampas (Viscera and muscle)ASE and SE ‡‡ (0.03 M AA)Acquity UPLC BEH HILICA: 5% ACN
B: 95% ACN + 1% AA pH 3.5
LC-MS/MS6For Solvent Std LOD: 0.074 ng/mL
LOQ: 0.123 ng/mL
For Matrix-matched Std LOD: 7.3 µg/kg LOQ: 24.5 µg/kg at S/N = 3 and 10
5–500 ng/mL (Solvent Std)
50–3000 µg/kg (Matrix-matched Std)
[48]
Charonia lampas lampas (Digestive gland)NMNMNMLC-MS/MS6, 22NRNR[45]
Gastropod
Nassarius spp.[28][28][28]HPLC-MSn (Ion trap) and HPLC-FLD4, 6, 7, 16, 22NRNR[28]
Gibbula umbilicalis, Monodonta lineata and Charonia lampas1% AA, DCM Defat *, C18 SPEXBridge™ Amide (LC- MS/MS)
Waters Acquity UPLC BEH Amide (UPLC-MS/MS)
For both,
A: 10 mM FA + 10 mM AF in H2O
B: 2 mM FA + 5 mM AF in ACN: H2O, 95:5
LC-MS/MS
UPLC-MS/MS
6, 7, 11/12, 14, 17/18, 22For LC- MS/MS
LOD: 16 ng/mL at S/N > 3
LOQ: 63 ng/mL S/N > 10
For UPLC-MS/MS
LOD: 1.7 ng/mL at S/N > 3
LOQ: 5 ng/mL S/N > 10
50–2000 ng/mL (LC-MS/MS)
31.25–3000 ng/mL (UPLC-MS/MS)
[47]
Grey side-gilled sea slug, Pleurobranchaea maculata
(Whole body)
50% MeOH, Strata Phenomonex SPETSK-GEL amide 80A: 10% ACN + (90% 50 mM FA + 2 mM AF in H2O)
B: 90% ACN + (10% 50 mM FA + 2 mM AF in H2O)
LC-MS/MS4, 6, 12, 16, 17/18LOD: 5 ng/mL (S/N = 50)5–250 ng/mL[73]
Blue-ringed octopus
Hapalochlaena fasciata and H. lunulata0.05 N AASynergi 4 µ Hydro-RP 80A C180.97% Heptafluorobutyric acid + 0.29% AA in 3% ACN (pH adjusted to 5.0 with NH4OH)Q-TOF MS6NR500 ng/mL to 0.5 mg/mL[76]
Blue-ringed octopus (Hapalochlaena fasiata and H. lunulata) (Posterior salivary gland, arm, dorsal mantle, ventral mantle, anterior salivary gland, digestive gland, testes conts./egg/paralarva, oviducal gland, brachial heart, nephridia, gill)0.05 N AASynergi 4 µ Hydro-RP 80A C183% ACN + 0.97% HFB + 0.29% AA, pH 5 LC-FLD6NR500 ng/mL–0.5 mg/mL[178]
Note: (Analyte*) Refer to Table 3 for the name of the TTX analytes.
Table 6. LC-MS methods for newts.
Table 6. LC-MS methods for newts.
SpeciesExtractionColumnMobile phaseMS Analytes *LOD and LOQLinear RangeReference
Cynops ensicauda popei (Whole body)O.2 M AA, Hexane Defat *, CharAd , Bio-Rex 70 and Hitachi gel 3011C SPETSK gel G1000PW (HILIC)16 mM AF, pH 5.5 + ACN (3:7, v/v)LC-MS/MS6, 7, 8, 11, 12, 14, 17, 19, 20, 23, 25, 27NRNR[24,26,59,175]
Notophthalmus viridescens (Whole body)0.1% AA + 70% EtOH, CharAd Develosil C30 UG-51% ACN + 20 mM
AHB + 10 mM AF, pH 4.0
LC-FLD4, 6, 8NRNR[37]
Notophthalmus viridescens (Whole body, liver and skin)0.1% AA in 70% MeOHDevelosil C30 UG-51% ACN + 30 mM AHB + 10 mM AF, pH 5.0LC-FLD6, 7, 8, 14, 15, LOD 0.4 pmol50–1000
pmol
[57,149]
Triturus spp. (Whole body)0.1% AA in 70% MeOH, CharAd Develosil (C30-UG-5)30 mM AHB + 10 mM AF, pH 5LC-FLD6, 8LOD
100 ng/g (TTX)
40 ng/g (6-epi TTX)
NR[35]
Taricha granulosa (Skin)0.1 M AASynergi 4 µ Hydro-RP 80A50 mM Am-acetate + 60 mM AHB, pH 5 in 1% ACNLC-FLD6NRNR[54,176]
Notophthalmus viridescens (Whole body)1% AA in 70% MeOHDevelosil C30-UG-51:11 vol.% ACN, 30 mM AHB + 10 mMAF, pH 5.0LC-FLD4, 6, 7, 8, 14, 15NRNR[53]
Taricha granulosa (Skin)0.1 M AASynergi 4 µ Hydro-RP 80A50 mM Am-acetate + 60 mM AHB, pH 5 in 1% ACNLC-FLD6, 7, 14NRNR[51]
Cynopus ensicauda (Skin)0.05 M AA, CharAd C30- UG-5 (LC-MS/MS and LC-FLD)20 mM AHB + 10 mM AF in 1% ACN, pH 4 (LC-MS/MS)
30 mM AHB + 10 mM AF in 1% ACN, pH 5 (LC-LFD)
LC-MS/MS
LC-LFD
6, 7, 8, 11, 12, 14, 15, 17, 18LOD: 0.7 pmol at S/N 250–1000 pmol[149]
Notophthalmus viridescens (Whole body)1% AA in 70% MeOHDevelosil C30-UG-5 (HPLC-FLD and LC-MS)30 mM AHB in 1% ACN, pH 5 (HPLC-FLD) 20 mM AHB + 10 mM AF in 1% ACN, pH 4 (LC-MS)HPLC-FLD
LC-MS
6, 7, 8, 11, 12, 14, 17, 18NRNR[50]
Cynops pyrrhogaster (Whole body)0.1% AAPuresil C1830 mM HFB + 1 mM Am-acetate, pH 5LC-MS6, 7, 8, 15NRNR[52]
Note: (Analyte*) Refer to Table 3 for the name of the TTX analytes.
Table 7. LC-MS methods for TTX and its analogues from crab and frog.
Table 7. LC-MS methods for TTX and its analogues from crab and frog.
SpeciesExtractionColumnMobile phaseMSAnalytes *LOD and LOQLinear RangeReference
Crabs
Demania cultripes, D. toxica, D. reynaudi, Lophozozymus incisus, L.pictor and Atergatopsis germaini (Appendage, cephalo-thorax and viscera)1%AA in MeOH, C18 cartridgeODS-330 mM HFB + 1 mM Am-acetate, pH 5LC-MS6, 7, 16LOD: 0.005 µg/mL0.03–3 µg/mL[52,139,]
Xanthias lividus (Appendage, cephalothorax and viscera)1%AA in MeOH, DCM Defat *, Bio-Gel P-2 filtration[67][67]HPLC6, 16NRNR[67]
Frogs
Brachycephalus ephippium, B. nodoterga and B. pernix (Whole Body, skin, liver and ovary)MeOH:AA (96:4), Amberlite GC-50 SPE, CharAdCLC-ODS
(LC-FLD)
Puresil C18
(LC-MS/MS)
0.06N HFB + 0.001N Am-acetate, pH 5 (CLC-ODS)
30 mM HFB + 1 mM Am-acetate, pH 5 (Puresil C18)
LC-FLD
LC-MS/MS
4, 6, 7, 8, 9, 11, 12, 14, 17NRNR[33,52,72,176]
Brachycephalus ephippium (Skin)1%AA in MeOH, Petroleum ether Defat*, CharAdCLC-ODS
(LC-FLD)
Puresil C18 (LC-MS/MS)
0.06N HFB + 0.001N Am-acetate, pH 5 (CLC-ODS)
30 mM HFB + 1 mM Am-acetate, pH 5 (Puresil C18)
LC-FLD
LC-MS/MS
4, 6, 7, 8, 9, 14NRNR[33,34,52,176]
Polypedates sp. (Skin, muscle and viscera)80% EtOH, pH 2, DCM Defat*, CharAd, 1% AA in 20% EtOH, Bio-Gel P2 and Bio-Rex 70 filtrationInertsil ODS-360 mM (NH4)3PO4, pH 5 + 10 mM HSA in 2% ACN LC-FLD6, 8, 14, 15NRNR[71]
Note: (Analyte*) Refer to Table 3 for the name of the TTX analytes.
Table 8. LC-MS methods for TTX and its analogues from bacteria.
Table 8. LC-MS methods for TTX and its analogues from bacteria.
SpeciesExtractionColumnMobile phaseMSAnalytes *LOD and LOQLinear RangeReference
Aeromonas strain from ovary of puffer fish, Takifugu obscurus0.1% AA, CharAd , Bio-Gel P2 and C18 SPEACQUITY UPLC BEH HILICA: 0.2% FA in H2O
B: 0.2% FA in ACN
Q-TOF MS6NR0–250 ng/mL[88]
Shewanella woodyi and Rosebacter sp. from copepod, Pseudocaligus fugu; ectoparasite of puffer fish, Takifugu pardalis0.1% AA, C18 SPE, CharAd [85,169]Asakawa et al. 2003 and Ito et al. 2006Asakawa et al. 2003 and Ito et al. 20066, 7, 16NRNR[85,169]
Vibrio strain, LM-1 from the intestine of puffer fish, Fugu vermicularis radiatesDCM Defat *, 0.03 M AA, Bio-Gel P-2 filtrationYMC-pack AM-314 octyldecyl silane0.05M HSA + 0.05M KH2PO4, pH 7 in MeOHLC-FLD6, 7, 16NRNR[91]
Nocardiopsis dassonvillei from the ovary of puffer fish, Fugu rubripes0.1% AA, CharAd , Bio-Gel P2 and Bio-Rex 70 filtrationBio-Rex 70MeOHLC-MS6NRNR[82,92]
Note: (Analyte*) Refer to Table 3 for the name of the TTX analytes.
Table 9. LC-MS methods for TTX and its analogues from human urine and blood.
Table 9. LC-MS methods for TTX and its analogues from human urine and blood.
SampleExtractionColumnMobile phaseMSAnalytes *LOD and LOQLinear RangeReference
Postmortem whole bloodMeOH, SPEPC(Phosphorychloline)–HILIC1% AA + ACN in MeOHLC-MS/MS6 and vogliboseLOD: 0.32 ng/mL
LOQ: 1.08 ng/mL
2–1200 ng/mL[104]
Urine and plasma2% AA, C18 and ZIC-HILIC SPEAtlantics dC1810 mM AF + FA (95:5, v/v) + 5 mM HFB in 2% ACNLC-MS/MS6LOD:
LOQ:
10–500 ng/mL[135]
Blood and urineC18 and Oasis MCX SPEAllsphere ODS-2 (LC-UV)
Nova-Pak C18 (LC-LFD)
Zorax 300SB-C3 (LC-MS/MS)
HILIC (LC-MS/MS)
Atlantics dC18 (LC-MS/MS)
4.8 mM 1-HSA +
41.8 mM SDP + 10% MeOH, pH (Allsphere ODS-2)
5 mM PIC B7 (HSA) + 3% MeCN in H2O, pH 4.5 (Nova-Pak C18)
10 mM TMA, 10 mM AF in 1% ACN, pH 4 (Zorax 300SB-C3)
0.1% FA in MeOH (HILIC)
10 mM AF + FA, (95:5, v/v) + 5 mM HFB + 2% ACN
LC-UV
LC-LFD
LC-MS/MS
LC-MS/MS
LC-MS/MS
6LOD: 10 ng/mL (LC-UV)
LOQ: 5 and 20 ng/mL for serum and urine (LC-LFD)
LOD: 15.6 nM (LC-MS/MS)
LOD: 0.1 ng/mL
LOQ: 1 ng/mL (LC-MS/MS)
LOD: 0.13 ngmL−1
LOQ: 2.5 ngmL−1
for urine and plasma
10–50,000 ng/mL (LC-UV)
20–300 for urine and 5–20 ng/mL for serum (LC-LFD)
93.75–9375 nM (LC-MS/MS)
1–100 ng/mL
0–500 ngmL−1 for urine and 0–20 ngmL−1 for plasma
[13,106,135,196]
Cooked and raw puffer fish (liver) and human urine1% AA in MeOHTosoHaas TSK-GEL Amide-805 mM AF + 26.5 mM FA in ACN: H2O, 70:30 LC-MS/MS620 µg/100g tissue1–10,000 ng/mL[136]
Urine and blood0.5 M AA, C18 SPEZorax 300SB-C31% ACN + 10 mM TMA + 10 mM AF, pH 4LC-MS6LOD: 15.6nM93.75–9375 nM[106]
Std mixtureNot usedTSKgel Amide-8016 mM AF, pH 5.5 + ACN (3:7, v/v)LC-MS/MS6, 7, 14, 22NR64 pg–2 ng
64 pg–2 ng
128 pg–1 ng
180 pg–1.4 ng
[128]
Serum0.5 M AA in MeOH, Oasis MCX SPECosmosil HILIC 4.6 × 150 mm 0.1% FA in water + MeOHLC-MS/MS (M. Horie et al., 2002)6, 7, 16LOD: 0.1 ng/mL
LOQ: 1 ng/mL
1–100 ng/mL[13,100]
Urine and serumUrine Extraction: C18 Sep-Pak SPE (0.2 M HCl in 20% MeOH) followed by Strata X-C 33 µm Cation Mixed-Mode Polymer SPE (0.1 M HCl+MeCN+MeOH+Water)
Serum Extraction: Oasis MCX SPE (0.2 M HCl in 20% MeOH +MeCN+MeOH+Water)
Nova-Pak C18 4 µm, 8 × 100 mmPIC B7 (Heptane sulfonic acid), 5 mM + 3% MeCN, pH 4.5 (adjusted with conc. NH3)LC-FLD6LOD:
20 ng/mL (Urine)
5 ng/mL (Serum)
LOQ:
20 ng/mL (Urine)
5 ng/mL (Serum)
20–300 ng/mL (Urine)
5–20 ng/mL (Serum)
[100]
Note: (Analyte*) Refer to Table 3 for the name of the TTX analytes.
Table 10. Recovery of TTX from different matrices.
Table 10. Recovery of TTX from different matrices.
MatrixExtraction Method% RecoveryReference
Trumpet shellASE (Accelerated solvent extraction) and SE (Solvent Extraction) (0.03 M AA) (UPLC–MS/MS)80–92[48]
Gastropod tissue1% AA in MeOH, C18-SPE, ultrafiltration (<3000 MW), (HPLC-FLD)90[13]
Xanthid crab, Demania cultripes1%AA in MeOH, C18-SPE, ultrafiltration (<3000 MW), (LC-MS)86.3 ± 2.9[139]
Puffer fish ovary0.05M AA, ODS-SPE, ultrafiltration (0.22 µ), (LC-MS)94.2–9108.3[127]
Puffer fish tissues, Muscle, Skin and Liver2% AA, methacrylate-styrene divinyl benzene cartridge (LC-MS) C18 column (50 mm × 2.1 mm i.d.) using 10 mmol/L IPCC-MS7-methanol (65:35)
as the mobile phase at a flow rate of 0.2 mL/min
Muscle 79–83
Skin 85–88
Liver 85–90
(LOD 0.01 µg/g tissue)
[244]
Puffer fish eggs and newt0.1% AA, Cosmosil 75C18-OPN resin-SPE, CHCl3 wash, (LC-MS)>90[175]
Newt (Whole body)0.1% AA in 70% MeOH, charcoal adsorption, (HPLC-FLD)50[35]
Blood serum0.5 M AA and Oasis MCX-SPE, ultrafiltration (<3000 MW) (LC-MS/MS)>95[13]
Whole Blood1% AA in MeOH, PCX-SPE, (LC-MS/MS)TTX 61.4Voglibose 62.8[104]
Human urine and plasmaC-18 and HILIC SPE (LC-MS/MS)75–81[135]
Human urine and blood0.5 M AA, C18 SPE, ultrafiltration (<3000 MW), (LC-MS)Urine 90.9 ± 1.4
Blood 90.6 ± 0.2
[106]
Human urine and blood2% AA, methacrylate-styrene divinyl benzene cartridge (LC-MS) C18 column (50 mm × 2.1 mm i.d.) using 10 mmol/L IPCC-MS7-methanol (65:35)
as the mobile phase at a flow rate of 0.2 mL/min
Human serum 93–96 (0.1 ng/mL)
Human urine 93-101 (0.1 ng/mL)
[244]
Combined muscle, liver and ovary from tiger puffers and muscle and ovary from balloon fishes1% AA in MeOH, defatted with chloroform (HPLC-FLD)91.0 ± 5.2[43]
Puffer fish muscle, liver and phosphate buffered saline1% AA in MeOH, defatted with chloroform (HPLC-FLD)86.4 ± 18.9[136]
Several extraction studies have been conducted to improve the recoveries of TTX in a number of sample types. Fong et al., 2011 [135] stated that acetonitrile and methanol give similar recoveries and in view of the global shortage of acetonitrile and consequently its increased expense the preferred choice must be methanol. The same group found that non-acidified solutions tend not to retain TTX so they used 2% acetic acid to enhance retention. In most of the studies conducted, scientists have used between 0.1 to 1% acetic acid for the extraction of TTX from different types of matrices (Table 4, Table 5, Table 6, Table 7, Table 8 and Table 9) and defatting of samples was usually accomplished with either chloroform, hexane or dichloromethane followed by charcoal adsorption for better recovery.
Many methods for the extraction of the TTXs include a solid phase extraction (SPE) step using C-18 cartridges and/or Bio-gel filtration for sample clean up prior to analysis. Recently, accelerated solvent extraction (ASE) was applied for the extraction of TTX from puffer fish and trumpet shell by Nzoughet et al., 2013 [48]. ASE gave better recoveries for the extraction of TTX from trumpet shell [48]. But the sample needs to be lyophilized (freeze-dried) which make it a time consuming process. Fong et al., 2011 [135] state that HILIC SPE should be carried out under gravity flow as the application of a vacuum compromises the recovery of TTX.

11.3.2. Development in Chromatography

Reverse phase chromatography was used for many years for the analysis of TTX and its analogues (most commonly C18). But all the analogues of TTX could not be separated using reverse phase chromatography. Others tried normal phase chromatography for the separation of the TTXs (most commonly TSK-GEL Amide-80 and HILIC). TTX is a polar compound and thus it retains quickly on reverse phase columns, but retains slowly on normal phase columns, also giving better separation of its analogues.
HILIC (Hydrophilic interaction liquid chromatography) column has hydrophilic stationary phase operated using reversed-phase type eluents; while ZIC-HILIC (Zwitter ionic hydrophilic interaction liquid chromatography) column has densely bonded, zwitter ionic functional groups. Separation is achieved by hydrophilic partitioning combined with weak ionic interactions for maximum selectivity, high load ability and easy optimization of methods. ZIC-HILIC columns are very efficient to improve peak resolution for polar and hydrophilic compounds.
Nakagawa et al., 2006 gave a brief description of the development of LC-MS for the detection of the TTXs [175]. They detected 11-deoxyTTX and 5,6,11-trideoxyTTX using a HILIC chromatography [175].
Chen et al., 2011 developed and validated a method for separation of TTX analogues from puffer fish [127]. They used an Atlantis HILIC Silica column (100 × 2.1 mm, 3 µm) with which they achieved a total run time of 10 min. Yotsu-Yamashita et al., 2011 could separate TTX from 4-epiTTX using a HILIC column. However 4,9-anhydro-5,6,11-trideoxyTTX and 4,4a-anhydro-5,6,11-trideoxyTTX were not clearly separated by HILIC chromatography [128]. In addition 5,6,11-trideoxyTTX and 8-epi-5,6,11-trideoxyTTX were not resolved using a HILIC chromatography, their determination was hindered from not only from sharing the same retention time but also by producing an identical MS/MS fragmentation pattern [59]. Of course the elution profile of the analogues of TTX on polar chromatography is opposite to that found in reverse phase chromatography [29]. A very good separation of TTX and its analogues was achieved using a ZIC-HILIC (5 µm, 150 × 2.1 mm) column from the extracts of Takifugu oblongus [138].
The most frequently applied mobile phases for TTX analysis were 10 mM ammonium formate + 10 mM formic acid in H2O and acetonitrile; 5 mM ammonium heptafluorobutyrate + 50 mM ammonium acetate or ammonium formate, pH 5 in 3% acetonitrile for puffer fish, gastropod, newt, bacteria and human urine and blood; 2 mM sodium 1-heptane sulfonate in 1% Methanol + K3PO4, 0.05 M, pH 7 and 3% Acetonitrile + 0.97% heptafluorobutyric acid + 0.29% AA, pH 5 for gastropod and newt. And 30 mM heptafluorobutyric acid + 1 mM ammonium acetate was often used as the mobile phase for the analysis of TTX in crab and frog samples.
The LC-MS methods that have been used for TTX analysis have been reviewed comprehensively by Leung et al., 2011 (they determined TTX levels using LC-MS in the urine and plasma of Asian patients) [196]. They brought total chromatographic run time to 5.5 min using an Atlantics dC18 (2.1 mm × 150 mm, 5 µm) column with flow rate of 200 µL min−1. This method was validated and applied on human urine and blood matrices for the detection of TTX [135]. They also studied the effect of an ion pair reagent (heptafluorobutyric acid) and optimized the concentration of the ion pair reagent at 5 mM. They found significant ion suppression with the MCX-HILIC, MAX-HILIC and HLB-HILIC columns but not with the C18-HILIC and Sep-Pak-HILIC. Finally they used C18-HILIC as it gave the most intense peaks.
The best reported LODs for TTX to-date are 0.074 ng/mL (0.23 nM) [48] in puffer fish and trumpet shell samples; 0.7 pmol (0.00022 ng/mL) at S/N 2 [149] in puffer fish samples; 0.4 pmol (0.00013 ng/mL) in newt samples [149]; 0.005 µg/mL (15.67 nM) in crab samples [139] and 0.1 ng/mL (0.31 nM) in human urine and plasma samples [13].

11.3.3. Development in Mass Spectrometry

The fragmentation patterns of TTX had been studied extensively by many research groups. However the fragmentation patterns of the analogues of TTX have not been not widely investigated. TTX gives characteristic fragments: m/z 302 (representing the loss of 1 water molecule), 284 (representing the loss of 3 water molecules), and at m/z 256, 178 and 162 [27,29,34,45,128,149]. 6-epiTTX gives characteristic fragments at m/z 302, 284, 256, 178 and 162 [149]. 4-epiTTX, 11-norTTX-6-(S)-ol, 4,9-anhydroTTX, 5-deoxyTTX and 5,6,11-trideoxyTTX have a common fragment at m/z 162 [128,149]. 11-norTTX-6-(S)-ol and 11-norTTX-6,6-diol both give a fragment at m/z 178 [149]. 5-deoxyTTX and 11-deoxyTTX give a characteristic fragment at m/z 176 [149]. 5-deoxyTTX and 5,6,11-trideoxyTTX give characteristic fragment at m/z 146 [149]. 11-oxoTTX gives a characteristic fragment at m/z 336, 318 (loss of 1 water molecule), 300 (loss of 2 water molecules), 282 (loss of 3 water molecules), and at m/z 178 and 162 [34].
Rodriguez et al., 2008 [45] suggests that the formation of the TTX MS ion at m/z 256 could be due to elimination of CO at C-10 causing cleavage of the bond between C-9 and C-10, C-10 and C-5-O and C-10 and C-7-O. The group also suggest that the formation of ion at m/z 254 in 5,6,11-trideoxy-TTX is due to the loss of one water molecule from the original structure. Fragment ions at m/z 162 and 178 are assigned as 2-aminohydroxyquinazoline and 2-aminodihydroxyquinazoline respectively which may be formed by the breaking of bonds between C-8a and C-9 and between C-6 and C-11 [149].
Some researchers have discovered unknown compounds that possess similar fragmentation patterns to TTX which may suggest the presence of unknown analogues of TTX in species known to contain TTX. Pires Jr. et al., 2005 [72] have found unknown compounds in Brachycephalus sp. with fragment ions at m/z 330 and m/z 348 which are also present in the MS/MS spectra of TTX; they have also identified ions at m/z 162 (2-aminohydroxyquinazoline) and m/z 178 (2-aminodihydroxyquinazoline) in those unknown compounds. There is wide scope for further investigations of fragmentation patterns of TTX analogues. Some of the analogues of TTX share the same mass and fragmentation pattern (two most intense ions) suggesting the need for full chromatographic resolution in studies relating to TTX.

11.3.4. Quantitative TTX Analysis

In most of the TTX studies, TTX analogues were quantified against a standard calibration curve of TTX, because certified standards for the analogues of TTX are commercially unavailable. In some studies, concentrations of TTX analogues have been calculated against standard calibration curves of individual analogues: for 4-epi TTX, 4,9-anhydro TTX and 5,6,11-deoxyTTX with good correlation regression coefficents (r2 = 0.99) obtained [128]. Kudo et al., 2012 calculated the concentration of 5,6,11-trideoxyTTX in samples against a standard curve of purified 5,6,11-trideoxyTTX [59].
In many studies TTX itself was used as the internal standard for the quantitation of its analogues [127]. Cho et al., 2012 [104] has used voglibose (m/z 268/92) as an internal standard for TTX (Figure 6). They used voglibose as an internal standard because isotope labelled TTX was not commercially available. Voglibose has a similar structure that of TTX and also had similar chromatographic and MS properties, so it is a good IS for TTX analysis. Man et al., 2010 [137] used salicylic acid as the internal standard.
Figure 6. Structure of voglibose (Internal standard for TTX).
Figure 6. Structure of voglibose (Internal standard for TTX).
Toxins 06 00693 g006

11.3.5. Matrix Effect in Puffer Fish, Trumpet Shell and Human Urine/Blood Sample

Ion suppression or matrix effects in LC-MS and LC-MS/MS is the reduction in detector response, or signal: noise produced by analytes of interest due to the competition for ionisation, between the analyte of interest and other compounds present in the sample matrix (e.g., polymers from plastic tubes and filters used for extraction, mobile phase additives etc.) which have not been removed from the sample matrix during extraction. Ion suppression effect reduces sensitivity of analysis if ion suppressing compounds co-elute with the analytes of interest. Therefore, it is very important to remove all the impurities from sample matrix [245].
Matrix effects in puffer fish, monkfish and human urine samples were compared between SPR and LC-MS/MS methods, however much of the detail was omitted from the publication [136] Tsai et al., 2006 [106] found that the levels of TTX were higher in urine than in blood samples 10 h after TTX ingestion. So urine is a better choice of sample for confirming TTX poisoning. Urine is a complex matrix containing many polar compounds therefore when using HILIC chromatography ion suppression caused by matrix components may compromise quantitation [196]. The ion suppression effect (post column infusion study), precision and accuracy (n = 10) was conducted for urine and plasma samples by Fong et al., 2011 [135]. They used double SPE (C-18 followed by ZIC-HILIC SPE) for better recovery of TTX. Cho et al., 2012 [104] obtained 98.3%–111.2% average matrix effect in blood when using the internal standard, voglibose (Figure 6) for their study.

12. Measures to Ensure Human Safety (Legislation)

Table 11 summarizes toxicity limits of TTX and some of its analogues. Acceptability limit in puffer fish as food in Japan is 10 MU TTX eq/g or 2.2 µg TTX eq/g of puffer flesh [131]. Regulatory limit for TTX in food is 2000 µg/kg TTX eq [246] while in the US a zero level of TTX in food exists [247]. But as it is newly emerging toxin in Europe, regulatory limits for TTX in food are still not established. There is a need of setting proper regulatory limits for TTX to ensure food safety.
Table 11. Toxicity limits for TTX.
Table 11. Toxicity limits for TTX.
DescriptionValueReference
Human median lethal dose8.7 µg/kg[127]
MLD for mammals (IP or IV)2.7–10 µg/kg for rats
4.5 µg/kg for guinea pigs
8–10 µg/kg for mice, rabbits, dogs and cats
[97]
Lethal potency5,000–6,000 MU/mg[46]
MLD for human10,000 MU (≈2 mg)[46]
Regulatory limit in food in USZero[247]
LD50 of TTX in mice9 µg/kg[19]
LD99 of 5,6,11-trideoxy TTX750 µg/kg [124]
IC50 for nine human functional voltage-gated Na+ channels≥1 μM[97]
MLD of TTX to humans2 mg/50 kg BW[131]
Minimum acute dose of TTX to humans0.2 mg/50 kg BW[131]
Acceptability limit in puffer fish as food in Japan10 MU TTX eq/g or
2.2 µg TTX eq/g of puffer flesh
[131]
LD50 of TTX in mice, dogs and rabbits8–14 µg/kg by injection[76]
Lethal dose2 mg[25]
LD50 of TTX10 µg/kg (IP in mice)[129]
LD50 of 11-deoxy TTX70 µg/kg (IP in mice)[31]
IC50 for 6,11-dideoxy TTX420 µg/kg (IP in mice)[125]
Regulatory limit in food in Japan2000 µg/kg TTX eq[246]
Lethal doses in KM miceLD1: 9.4 µg/kg
LD50: 11.3 µg/kg
LD99: 13.5 µg/kg
[130]

13. Conclusions

In areas where TTX occurs with regularity, it is important that rapid analytical methods are deployed for the analysis of clinical samples, most especially blood and urine in suspected poisoning victims. LC-MS/MS methodologies are particularly appropriate to detect TTX and its analogues in clinical samples with the speed required in such cases. Though there is still no commercially available antidote to TTX, it may dictate the course of medical treatment, especially for those with compromised renal function.
There is hope that in the future an antidote may be developed to counteract the effects of the toxin in vivo. In the meantime more research is required on the prevalence and the toxicity of TTX metabolic bioconversion products in vector species in order to develop a comprehensive human risk analysis.
A report in the wildlife section of “The Times” newspaper dated 11 May 2013 [248] revealed that a red scorpion fish had been caught in the Celtic Sea off the coast of Ireland and England. This is clear evidence that exotic and toxic marine species can travel and may be commonly found in cooler European waters in the future. If migration trends like this are to continue and there are predictions that they are likely (in view of global warming), it may be prudent to carry out surveillance of susceptible marine species, algae and seawater in European territory for TTX and other toxins associated with warmer regions.

Acronyms

4-AP
4-aminopyridine
AA
Acetic acid
ACN
Acetonitrile
AF
Ammonium formate
AHB
Ammonium heptafluoro butyric acid
Am-acetate
Ammonium acetate
Am-OH
Ammonium hydroxide
ASE
Accelerated solvent extraction
BW
Body weight
CharAd
Charcoal adsorption
CID
Collision induced dissociation
DCM
Dichloromethane
Defat *
Defatting
ELISA
Enzyme linked immunosorbent assay
Eq
Equivalent
EtOH
Ethanol
FA
Formic acid
GC-MS
Gas chromatography- Mass spectrometry
GTX
Gonyautoxin
HCD
High collision induced dissociation
HFB
Heptafluoro butyric acid
HLB-HILIC
Hydrophilic-lipophilic balance- HILIC
HILIC
Hydrophilic interaction liquid chromatography
HPLC
High performance liquid chromatography
HAS
Heptane sulfonic acid
IC50
Half maximal inhibitory concentration is a measure of the effectiveness of a compound in inhibiting biological or biochemical function
IP
Intraperitoneal
IR
Infrared resonance
IV
Intravenous
LC-FLD
Liquid chromatography-fluorescent detection
LC-MS
Liquid chromatography-Mass spectrometry
LC-PDA
Liquid chromatography-Photo diode array detection
LC-UV
Liquid chromatography-Ultra violet detection
LD50
Lethal dose50 of a toxin is the dose required to kill 50% of the members of a tested population after a specified test duration.
LD99
Lethal dose99 of a toxin is the dose required to kill 99% of the members of a tested population after a specified test duration.
LOD
Limit of detection
LOQ
Limit of quantitation
MAX-HILIC
Mixed-mode anion exchange- HILIC
MCX-HILIC
Mixed-mode cation exchange- HILIC
MeCN
Methyl cyanide
MeOH
Methanol
MLD
Median lethal dose
MRM
Multiple reaction monitoring
MU
Mouse unit; 1 MU is defined as the amount of toxin required to kill a 20 g ICR (Institute of Cancer Research) strain mouse in 30 min after intraperitoneal injection [81]
MW
Molecular weight
m/z
mass/charge ratio
NMR
Nuclear magnetic resonance
PSP
Paralytic shell fish poisoning
Q-TOF MS
Quadrupole-time-of-flight mass spectrometry
SE
solvent extraction
SIR
Selected ion recording
S/N
Signal/Noise ratio
SPE
Solid phase extraction
STX
Saxitoxin
TBA-PO4
Tetrabutyl ammonium phosphate
TLC
Thin layer chromatography
TMA
Trimethyl amine
Tris-Ac
Tris- acetate buffer
TTX
Tetrodotoxin
UPLC
Ultra performance liquid chromatography
ZIC-HILIC
Zwitter ionic hydrophilic interaction liquid chromatography

Acknowledgments

The authors acknowledge the funding obtained under the EU/INTERREG IIIB Atlantic Area Programme for projects titled ATLANTOX “Advanced Tests about New Toxins Occurring in the Atlantic Area due to Climate Change” and PHARMATLANTIC “Knowledge Transfer Network for Prevention of Mental Diseases and Cancer in the Atlantic Area”. The Higher Education Authority (Programme for Research in Third-Level Institutions, Cycle 4 (PRTLI IV) National Collaboration Programme on Environment and Climate Changes: Impacts and Responses is also acknowledged for funding marine research within CIT.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kan, S.K.; Chan, M.K.; David, P. Nine fatal cases of puffer fish poisoning in Sabah, Malaysia. Med. J. Malays. 1987, 42, 199–200. [Google Scholar]
  2. U.S. Department of Health and Human Services. Tetrodotoxin Poisoning Associated with Eating Puffer Fish Transported from Japan—California, 1996; Morbidity and Mortality Weekly Report; Centre For Disease Control and Prevention: Atlanta, GA, USA, 1996; Volume 45, pp. 389–412.
  3. Mahmud, Y.; Tanu, M.B.; Noguchi, T. First occurrence of a food poisoning incident due to ingestion of Takifugu oblongus, along with a toxicological report on three marine puffer species in Bangladesh. J. Food Hyg. Soc. Jpn. 1999, 40, 473–480. [Google Scholar] [CrossRef]
  4. Hwang, D.F.; Shiu, Y.C.; Hwang, P.A.; Lu, Y.H. Tetrodotoxin in gastropods (snails) implicated in food poisoning in Northern Taiwan. J. Food Prot. 2002, 65, 1341–1344. [Google Scholar]
  5. Sui, L.M.; Chen, K.; Hwang, P.A.; Hwang, D.F. Identification of tetrodotoxin in marine gastropods implicated in food poisoning. J. Nat. Toxins 2002, 11, 213–220. [Google Scholar]
  6. How, C.-K.; Chern, C.-H.; Huang, Y.-C.; Wang, L.-M.; Lee, C.-H. Tetrodotoxin poisoning. Am. J. Emerg. Med. 2003, 21, 51–54. [Google Scholar] [CrossRef]
  7. Hwang, P.-A.; Tsai, Y.-H.; Lu, Y.-H.; Hwang, D.-F. Paralytic toxins in three new gastropod (Olividae) species implicated in food poisoning in southern Taiwan. Toxicon 2003, 41, 529–533. [Google Scholar] [CrossRef]
  8. Shiu, Y.-C.; Lu, Y.-H.; Tsai, Y.-H.; Chen, S.-K.; Hwang, D.-F. Occurrence of Tetrodotoxin in the Causative Gastropod Polinices didyma and another Gastropod Natica lineata collected from western Taiwan. J. Food Drug Anal. 2003, 11, 159–163. [Google Scholar]
  9. Ahasan, H.A.M.N.; Mamun, A.A.; Karim, S.R.; Bakar, M.A.; Gazi, E.A.; Bala, C.S. Paralytic complications of puffer fish (tetrodotoxin) poisoning. Singap. Med. J. 2004, 45, 73–74. [Google Scholar]
  10. Hwang, P.A.; Tsai, Y.H.; Deng, J.F.; Cheng, C.A.; Ho, P.H.; Hwang, D.F. Identification of tetrodotoxin in a marine gastropod (Nassarius glans) responsible for human morbidity and mortality in Taiwan. J. Food Prot. 2005, 68, 1696–1701. [Google Scholar]
  11. Ahmed, S. Puffer fish tragedy in Bangladesh: An incident of Takifugu oblongus poisoning in Degholia, Khulna. Afr. J. Mar. Sci. 2006, 28, 457–458. [Google Scholar] [CrossRef]
  12. Jen, H.C.; Lin, S.J.; Lin, S.Y.; Huang, Y.W.; Liao, I.C.; Arakawa, O.; Hwang, D.F. Occurrence of tetrodotoxin and paralytic shellfish poisons in a gastropod implicated in food poisoning in southern Taiwan. Food Addit. Contam. 2007, 8, 902–909. [Google Scholar]
  13. Jen, H.-C.; Lin, S.-J.; Tsai, Y.-H.; Chen, C.-H.; Lin, Z.-C.; Hwang, D.-F. Tetrodotoxin poisoning evidenced by solid-phase extraction combining with liquid chromatography-tandem mass spectrometry. J. Chromatogr. B 2008, 871, 95–100. [Google Scholar] [CrossRef]
  14. Noguchi, T.; Arakawa, O.; Takatani, T. TTX accumulation in pufferfish—Review. Comp. Biochem. Physiol. D 2006, 1, 145–152. [Google Scholar]
  15. Saoudi, M.; Rabeh, F.B.; Jammoussi, K.; Abdelmouleh, A.; Belbahri, L.; Feki, A.E. Biochemical and physiological responses in Wistar rat after administration of puffer fish (Lagocephalus lagocephalus) flesh. J. Food Agric. Environ. 2007, 5, 107–111. [Google Scholar]
  16. Machida, S. Food poisoning due to ingestion of “ivory shell” Babylonia japonica. J. Food Hyg. Soc. Jpn. 1965, 6, 87–89. [Google Scholar]
  17. Behera, A.; Dash, B.K.; Barik, B.K. Rare puffer fish poisoning—A case report. Medico-Leg. Update 2008, 8, 5–6. [Google Scholar]
  18. Homaira, N.; Rahman, M.; Luby, S.P.; Rahman, M.; Haider, M.S.; Faruque, L.I.; Khan, D.; Parveen, S.; Gurley, E.S. Multiple outbreaks of puffer fish intoxication in Bangladesh, 2008. Am. J. Trop. Med. Hyg. 2010, 83, 440–444. [Google Scholar] [CrossRef]
  19. Islam, Q.T.; Razzak, M.A.; Islam, M.A.; Bari, M.I.; Basher, A.; Chowdhury, F.R.; Sayeduzzaman, A.B.M.; Ahasan, H.A.M.N.; Faiz, M.A.; Arakawa, O.; et al. Puffer fish poisoning in Bangladesh: Clinical and toxicological results from large outbreaks in 2008. Trans. R. Soc. Trop. Med. Hyg. 2011, 105, 74–80. [Google Scholar] [CrossRef]
  20. Suehiro, M. Historical review on chemical and medical studies of globefish toxin before World War II. Jpn. Soc. Hist. Pharm. 1994, 29, 428–434. [Google Scholar]
  21. Do, H.K.; Kogure, K.; Simidu, U. Identification of deep-sea-sediment bacteria which produce tetrodotoxin. Appl. Environ. Microbiol. 1990, 56, 1162–1163. [Google Scholar]
  22. Asakawa, M.; Toyoshima, T.; Shida, Y.; Noguchi, T.; Miyazawa, K. Paralytic toxins in a ribbon worm Cephalothrix species (Nemertean) adherent to cultured oysters in Hiroshima Bay, Hiroshima Prefecture, Japan. Toxicon 2000, 38, 763–773. [Google Scholar] [CrossRef]
  23. Mahmud, Y.; Okada, K.; Takatani, T.; Kawatsu, K.; Hamano, Y.; Arakawa, O.; Noguchi, T. Intra-tissue distribution of tetrodotoxin in two marine puffers Takifugu vermicularis and Chelonodon patoca. Toxicon 2003, 41, 13–18. [Google Scholar] [CrossRef]
  24. Jang, J.; Yotsu-Yamashita, M. Distribution of tetrodotoxin, saxitoxin, and their analogs among tissues of the puffer fish Fugu pardalis. Toxicon 2006, 48, 980–987. [Google Scholar] [CrossRef]
  25. Huang, H.-N.; Lin, J.; Lin, H.-L. Identification and quantification of tetrodotoxin in the marine gastropod Nassarius by LC-MS. Toxicon 2008, 51, 774–779. [Google Scholar] [CrossRef]
  26. Jang, J.-H.; Lee, J.-S.; Yotsu-Yamashita, M. LC/MS analysis of tetrodotoxin and its deoxy analogs in the marine puffer fish Fugu niphobles from the southern coast of Korea, and in the brackishwater puffer fishes Tetraodon nigroviridis and Tetraodon biocellatus from Southeast Asia. Mar. Drugs 2010, 8, 1049–1058. [Google Scholar] [CrossRef]
  27. Chulanetra, M.; Sookrung, N.; Srimanote, P.; Indrawattana, N.; Thanongsaksrikul, J.; Sakolvaree, Y.; Chongsa-Nguan, M.; Kurazono, H.; Chaicumpa, W. Toxic marine puffer fish in Thailand Seas and tetrodotoxin they contained. Toxins 2011, 3, 1249–1262. [Google Scholar] [CrossRef]
  28. Luo, X.; Yu, R.C.; Wang, X.J.; Zhou, M.J. Toxin composition and toxicity dynamics of marine gastropod Nassarius spp. collected from Lianyungang, China. Food Addit. Contam. A 2012, 29, 117–127. [Google Scholar] [CrossRef]
  29. Rodríguez, P.; Alfonso, A.; Otero, P.; Katikou, P.; Georgantelis, D.; Botana, L.M. Liquid chromatography-mass spectrometry method to detect Tetrodotoxin and Its analogues in the puffer fish Lagocephalus sceleratus (Gmelin, 1789) from European waters. Food Chem. 2012, 132, 1103–1111. [Google Scholar] [CrossRef]
  30. Kim, Y.H.; Brown, G.H.; Mosher, H.S.; Fuhrman, F.A. Tetrodotoxin: Occurrence in Atelopid frogs of Costa Rica. Science 1975, 189, 151–152. [Google Scholar]
  31. Yasumoto, T.; Yotsu, M.; Murate, M.; Naoki, H. New tetrodotoxin analogues from the newt Cynops ensicauda. J. Am. Chem. Soc. 1988, 110, 2344–2345. [Google Scholar] [CrossRef]
  32. Yotsu, M.; Iorizzi, M.; Yasumoto, T. Distribution of tetrodotoxin, 6-epitetrodotoxin, and 11-deoxytetrodotoxin in newts. Toxicon 1990, 28, 238–241. [Google Scholar] [CrossRef]
  33. Pires, O.R., Jr.; Sebben, A.; Schwartz, E.F.; Largura, S.W.R.; Bloch, C., Jr.; Morales, R.A.V.; Schwartz, C.A. Occurrence of tetrodotoxin and its analogues in the Brazilian frog Brachycephalus ephippium (Anura: Brachycephalidae). Toxicon 2002, 40, 761–766. [Google Scholar] [CrossRef]
  34. Pires, O.R., Jr.; Sebben, A.; Schwartz, E.F.; Bloch, C., Jr.; Morales, R.A.V.; Schwartz, C.A. The occurrence of 11-oxotetrodotoxin, a rare tetrodotoxin analogue, in the brachycephalidae frog Brachycephalus ephippium. Toxicon 2003, 42, 563–566. [Google Scholar] [CrossRef]
  35. Yotsu-Yamashita, M.; Mebs, D.; Kwet, A.; Schneider, M. Tetrodotoxin and its analogue 6-epitetrodotoxin in newts (Triturus spp.; Urodela, Salamandridae) from southern Germany. Toxicon 2007, 50, 306–309. [Google Scholar] [CrossRef]
  36. Mebs, D.; Yotsu-Yamashita, M. Tetrodotoxin in North-American newts. Toxicon 2012, 60, 1–120. [Google Scholar] [CrossRef]
  37. Yotsu-Yamashita, M.; Gilhen, J.; Russell, R.W.; Krysko, K.L.; Melaun, C.; Kurz, A.; Kauferstein, S.; Kordis, D.; Mebs, D. Variability of tetrodotoxin and of its analogues in the red-spotted newt, Notophthalmus viridescens (Amphibia: Urodela: Salamandridae). Toxicon 2012, 59, 257–264. [Google Scholar] [CrossRef]
  38. Noguchi, T.; Ebesu, J.S.M. Puffer poisoning: Epidemiology and treatment. Toxin Rev. 2001, 20, 1–10. [Google Scholar] [CrossRef]
  39. Saoudi, M.; Abdelmouleh, A.; El Feki, A. Tetrodotoxin: A potent marine toxin. Toxin Rev. 2010, 29, 60–70. [Google Scholar] [CrossRef]
  40. Narita, H.; Noguchi, T.; Maruyama, J.; Ueda, Y.; Hashlmoto, K.; Watanade, Y.; Hida, K. Occurrence of tetrodotoxin in a trumpet shell, “boshubora” Charonia sauliae. Bull. Jpn. Soc. Sci. Fish 1981, 47, 935–941. [Google Scholar] [CrossRef]
  41. Hwang, D.F.; Lin, L.C.; Jeng, S.S. Variation and secretion of toxins in gastropod mollusc Niotha clathrata. Toxicon 1992, 30, 1189–1194. [Google Scholar] [CrossRef]
  42. Hwang, D.F.; Cheng, C.A.; Tsai, H.T.; Shih, D.Y.C.; Ko, H.C.; Yang, R.Z.; Jeng, S.S. Identification of tetrodotoxin and paralytic shellfish toxins in marine gastropods implicated in food poisoning. Fish. Sci. 1995, 61, 675–679. [Google Scholar]
  43. Chen, C.-Y.; Chou, H.-N. Detection of tetrodotoxin by high performance liquid chromatography in lined-moon shell and puffer fish. Acta Zool. Taiwan 1998, 9, 41–48. [Google Scholar]
  44. Cassiday, L. First report of TTX in a European trumpet shell. Anal. Chem. 2008, 80. [Google Scholar] [CrossRef]
  45. Rodriguez, P.; Alfonso, A.; Vale, C.; Alfonso, C.; Vale, P.; Tellez, A.; Botana, L.M. First toxicity report of tetrodotoxin and 5,6,11-TrideoxyTTX in the trumpet shell Charonia lampas lampas in Europe. Anal. Chem. 2008, 80, 5622–5629. [Google Scholar] [CrossRef]
  46. Noguchi, T.; Onuki, K.; Arakawa, O. Tetrodotoxin poisoning due to pufferfish and gastropods, and their intoxication mechanism. ISRN Toxicol. 2011, 2011, 1–10. [Google Scholar] [CrossRef]
  47. Silva, M.; Azevedo, J.; Rodriguez, P.; Alfonso, A.; Botana, L.M.; Vasconcelos, V. New gastropod vectors and tetrodotoxin potential expansion in temperate waters of the Atlantic Ocean. Mar. Drugs 2012, 10, 712–726. [Google Scholar] [CrossRef]
  48. Nzoughet, J.K.; Campbell, K.; Barnes, P.; Cooper, K.M.; Chevallier, O.P.; Elliott, C.T. Comparison of sample preparation methods, validation of an UPLC-MS/MS procedure for the quantification of tetrodotoxin present in marine gastropods and analysis of pufferfish. Food Chem. 2013, 136, 1584–1589. [Google Scholar] [CrossRef]
  49. Kotaki, Y.; Shimizu, Y. 1-Hydroxy-5,11-dideoxytetrodotoxin, the first N-hydroxy and ring-deoxy derivative of tetrodotoxin found in the newt Taricha granulosa. J. Am. Chem. Soc. 1993, 115, 827–830. [Google Scholar] [CrossRef]
  50. Yotsu-Yamashita, M.; Mebs, D. The levels of tetrodotoxin and its analogue 6-epitetrodotoxin in the red-spotted newt, Notophthalmus viridescens. Toxicon 2001, 39, 1261–1263. [Google Scholar] [CrossRef]
  51. Hanifin, C.T.; BrodieIII, E.D.; Brodie, E.D., Jr. Tetrodotoxin levels of the rough-skin newt, Taricha granulosa, increase in long-term captivity. Toxicon 2002, 40, 1149–1153. [Google Scholar] [CrossRef]
  52. Tsuruda, K.; Arakawa, O.; Kawatsu, K.; Hamano, Y.; Takatani, T.; Noguchi, T. Secretory glands of tetrodotoxin in the skin of the Japanese newt Cynops pyrrhogaster. Toxicon 2002, 40, 131–136. [Google Scholar] [CrossRef]
  53. Yotsu-Yamashita, M.; Mebs, D. Occurrence of 11-oxotetrodotoxin in the red-spotted newt, Notophthalmus viridescens, and further studies on the levels of tetrodotoxin and its analogues in the newt’s efts. Toxicon 2003, 41, 893–897. [Google Scholar] [CrossRef]
  54. Cardall, B.L.; Brodie, E.D., Jr.; Brodie, E.D., III; Hanifin, C.T. Secretion and regeneration of tetrodotoxin in the rough-skin newt (Taricha granulosa). Toxicon 2004, 44, 933–938. [Google Scholar] [CrossRef]
  55. Zimmer, R.K.; Schar, D.W.; Ferrer, R.P.; Krug, P.J.; Kats, L.B.; Michel, W.C. The scent of danger: Tetrodotoxin (TTS) as an olfactory cue of predation risk. Ecol. Monogr. 2006, 76, 585–600. [Google Scholar] [CrossRef]
  56. Ferrer, R.P.; Zimmer, R.K. The scent of danger: Arginine as an olfactory cue of reduced predation risk. J. Exp. Biol. 2007, 210, 1768–1775. [Google Scholar] [CrossRef]
  57. Mebs, D.; Arakawa, O.; Yotsu-Yamashita, M. Tissue distribution of tetrodotoxin in the red-spotted newt Notophthalmus viridescens. Toxicon 2010, 55, 1353–1357. [Google Scholar] [CrossRef]
  58. Gall, B.G.; Stokes, A.N.; French, S.S.; Schlepphorst, E.A.; Brodie, E.D., 3rd.; Brodie, E.D., Jr. Tetrodotoxin levels in larval and metamorphosed newts (Taricha granulosa) and palatability to predatory dragonflies. Toxicon 2011, 57, 978–983. [Google Scholar] [CrossRef]
  59. Kudo, Y.; Yasumoto, T.; Konoki, K.; Cho, Y.; Yotsu-Yamashita, M. Isolation and structural determination of the first 8-epi-type tetrodotoxin analogs from the newt, Cynops ensicauda popei, and comparison of tetrodotoxin analogs profiles of this newt and the puffer fish, Fugu poecilonotus. Mar. Drugs 2012, 10, 655–667. [Google Scholar] [CrossRef]
  60. Mebs, D.; Yotsu-Yamashita, M.; Seitz, H.M.; Arakawa, O. Tetrodotoxin does not protect red-spotted newts, Notophthalmus viridescens, from intestinal parasites. Toxicon 2012, 60, 66–69. [Google Scholar] [CrossRef]
  61. Noguchi, T.; Jeon, J.K.; Arakawa, O.; Sugita, H.; Deguchi, Y.; Hashimoto, K. Occurrence of tetrodotoxin and anhydrotetrodotoxin in Vibrio sp. isolated from the intestines of xanthid crab Atergatis floridus. J. Biochem. 1986, 99, 311–314. [Google Scholar]
  62. Arakawa, O.; Noguchi, T.; Shida, Y.; Onoue, Y. Occurence of 11-oxotetrodotoxin and 11-nortetrodotoxin-6(R)-ol in a xanthid crab Atergatis floridus collected at Kojima, Ishigaki Island. Fish. Sci. 1994, 60, 769–771. [Google Scholar]
  63. Kanchanapongkul, J.; Krittayapoositpot, P. An epidemic of tetrodotoxin poisoning following ingestion of the horseshoe crab Carcinoscorpius rotundicauda. Southeast Asian J. Trop. Med. Public Health 1995, 26, 364–367. [Google Scholar]
  64. Hwang, D.F.; Tsai, Y.H. Toxicological study on the xanthid crab Zosimus aeneus in Taiwan. Toxicon 1997, 35. [Google Scholar] [CrossRef]
  65. Tsai, Y.-H.; Hwang, D.-F.; Chai, T.-J.; Jeng, S.-S. Toxicity and toxic components of two xanthid crabs, Atergatis floridus and Demania reynaudi, in Taiwan. Toxicon 1997, 35, 1327–1335. [Google Scholar] [CrossRef]
  66. Llewellyn, L.E.; Dodd, M.J.; Robertson, A.; Ericson, G.; de Koning, C.; Negri, A.P. Post-mortem analysis of samples from a human victim of a fatal poisoning caused by the xanthid crab, Zosimus aeneus. Toxicon 2002, 40, 1463–1469. [Google Scholar] [CrossRef]
  67. Tsai, Y.-H.; Ho, P.-H.; Jeng, S.-S.; Hwang, D.-F. Paralytic toxins in Taiwanese crab Xanthias lividus. Fish. Sci. 2002, 68, 659–661. [Google Scholar]
  68. Jester, R.; Rhodes, L.; Beuzenberg, V. Uptake of paralytic shellfish poisoning and spirolide toxins by paddle crabs (Ovalipes catharus) via a bivalve vector. Harmful Algae 2009, 8, 369–376. [Google Scholar] [CrossRef]
  69. Lin, H.; Nagashima, Y.; Jiang, P.; Qin, X.; Lu, Y.; Zhang, C. Screening for toxicity and resistance to paralytic shellfish toxin of shore crabs inhabiting at Leizhou peninsula, China. Mar. Environ. Res. 2012, 78, 48–52. [Google Scholar] [CrossRef]
  70. Yang, L.; Kao, C.Y. Actions of chiriquitoxin on frog skeletal muscle fibers and implications for the tetrodotoxin/saxitoxin receptor. J. Gen. Physiol. 1992, 100, 609–622. [Google Scholar] [CrossRef]
  71. Tanu, M.B.; Mahmud, Y.; Tsuruda, K.; Arakawa, O.; Noguchi, T. Occurrence of tetrodotoxin in the skin of a rhacophoridid frog Polypedates sp. from Bangladesh. Toxicon 2001, 39, 937–941. [Google Scholar] [CrossRef]
  72. Pires, O.R., Jr.; Sebben, A.; Schwartz, E.F.; Morales, R.A.V.; Bloch, C., Jr.; Schwartz, C.A. Further report of the occurrence of tetrodotoxin and new analogues in the Anuran family Brachycephalidae. Toxicon 2005, 45, 73–79. [Google Scholar] [CrossRef]
  73. McNabb, P.; Selwood, A.I.; Munday, R.; Wood, S.A.; Taylor, D.I.; MacKenzie, L.A.; van Ginkel, R.; Rhodes, L.L.; Cornelisen, C.; Heasman, K.; et al. Detection of tetrodotoxin from the grey side-gilled sea slug—Pleurobranchaea maculata, and associated dog neurotoxicosis on beaches adjacent to the Hauraki Gulf, Auckland, New Zealand. Toxicon 2010, 56, 466–473. [Google Scholar] [CrossRef]
  74. Wood, S.; Casas, M.; Taylor, D.; McNabb, P.; Salvitti, L.; Ogilvie, S.; Cary, S.C. Depuration of tetrodotoxin and changes in bacterial communities in Pleurobranchea maculata adults and egg masses maintained in captivity. J. Chem. Ecol. 2012, 38, 1342–1350. [Google Scholar] [CrossRef]
  75. Lin, S.-J.; Hwang, D.-F. Possible source of tetrodotoxin in the starfish Astropecten scoparius. Toxicon 2001, 39, 573–579. [Google Scholar] [CrossRef]
  76. Williams, B.L.; Caldwell, R.L. Intra-organismal distribution of tetrodotoxin in two species of blue-ringed octopuses (Hapalochlaena fasciata and H. lunulata). Toxicon 2009, 54, 345–353. [Google Scholar] [CrossRef]
  77. Williams, B.; Hanifin, C.; Brodie, E.; Caldwell, R. Ontogeny of tetrodotoxin levels in blue-ringed octopuses: Maternal investment and apparent independent production in offspring of Hapalochlaena lunulata. J. Chem. Ecol. 2011, 37, 10–17. [Google Scholar]
  78. Williams, B.L.; Stark, M.R.; Caldwell, R.L. Microdistribution of tetrodotoxin in two species of blue-ringed octopuses (Hapalochlaena lunulata and Hapalochlaena fasciata) detected by fluorescent immunolabeling. Toxicon 2012, 60, 1307–1313. [Google Scholar] [CrossRef]
  79. Ali, A.E.; Arakawa, O.; Noguchi, T.; Miyazawa, K.; Shida, Y.; Hashimoto, K. Tetrodotoxin and related substances in a ribbon worm Cephalothrix linearis (Nemertean). Toxicon 1990, 28, 1083–1093. [Google Scholar] [CrossRef]
  80. Cheng, C.A.; Hwang, D.F.; Tsai, Y.H.; Chen, H.C.; Jeng, S.S.; Noguchi, T.; Ohwada, K.; Hasimoto, K. Microflora and tetrodotoxin-producing bacteria in a gastropod, Niotha clathrata. Food Chem. Toxicol. 1995, 33, 929–934. [Google Scholar] [CrossRef]
  81. Yu, C.-F.; Yu, P.H.-F.; Chan, P.-L.; Yan, Q.; Wong, P.-K. Two novel species of tetrodotoxin-producing bacteria isolated from toxic marine puffer fishes. Toxicon 2004, 44, 641–647. [Google Scholar] [CrossRef]
  82. Wu, Z.; Yang, Y.; Xie, L.; Xia, G.; Hu, J.; Wang, S.; Zhang, R. Toxicity and distribution of tetrodotoxin-producing bacteria in puffer fish Fugu rubripes collected from the Bohai Sea of China. Toxicon 2005, 46, 471–476. [Google Scholar]
  83. Yan, Q.; Yu, P.H.-F.; Li, H.-Z. Detection of tetrodotoxin and bacterial production by Serratia Marcescens. World J. Microbiol. Biotechnol. 2005, 21, 1255–1258. [Google Scholar] [CrossRef]
  84. Croci, L.; Cozzi, L.; Suffredini, E.; Ciccaglioni, G.; Toti, L.; Milandri, A.; Ceredi, A.; Benzi, M.; Poletti, R. Characterization of microalgae and associated bacteria collected from shellfish harvesting areas. Harmful Algae 2006, 5, 266–274. [Google Scholar] [CrossRef]
  85. Venmathi Maran, B.A.; Iwamoto, E.; Okuda, J.; Matsuda, S.; Taniyama, S.; Shida, Y.; Asakawa, M.; Ohtsuka, S.; Nakai, T.; Boxshall, G.A. Isolation and characterization of bacteria from the copepod Pseudocaligus fugu ectoparasitic on the panther puffer Takifugu pardalis with the emphasis on TTX. Toxicon 2007, 50, 779–790. [Google Scholar] [CrossRef]
  86. Wang, X.J.; Yu, R.C.; Luo, X.; Zhou, M.J.; Lin, X.T. Toxin-screening and identification of bacteria isolated from highly toxic marine gastropod Nassarius semiplicatus. Toxicon 2008, 52, 55–61. [Google Scholar] [CrossRef]
  87. Wang, J.; Fan, Y. Isolation and characterization of a Bacillus species capable of producing tetrodotoxin from the puffer fish Fugu obscurus. World J. Microbiol. Biotechnol. 2010, 26, 1755–1760. [Google Scholar] [CrossRef]
  88. Yang, G.; Xu, J.; Liang, S.; Ren, D.; Yan, X.; Bao, B. A novel TTX-producing Aeromonas isolated from the ovary of Takifugu obscurus. Toxicon 2010, 56, 324–329. [Google Scholar] [CrossRef]
  89. Kono, M.; Matsui, T.; Furukawa, K.; Yotsu-Yamashita, M.; Yamamori, K. Accumulation of tetrodotoxin and 4,9-anhydrotetrodotoxin in cultured juvenile kusafugu Fugu niphobles by dietary administration of natural toxic komonfugu Fugu poecilonotus liver. Toxicon 2008, 51, 1269–1273. [Google Scholar] [CrossRef]
  90. Noguchi, T.; Arakawa, O.; Takatani, T. Toxicity of pufferfish Takifugu rubripes cultured in netcages at sea or aquaria on land. Comp. Biochem. Phys. D 2006, 1, 153–157. [Google Scholar]
  91. Lee, M.-J.; Jeong, D.-Y.; Kim, W.-S.; Kim, H.-D.; Kim, C.-H.; Park, W.-W.; Park, Y.-H.; Kim, K.-S.; Kim, H.-M.; Kim, D.-S. A tetrodotoxin-producing Vibrio strain, LM-1, from the puffer fish Fugu vermicularis radiatus. Appl. Environ. Microbiol. 2000, 66, 1698–1701. [Google Scholar] [CrossRef]
  92. Wu, Z.; Xie, L.; Xia, G.; Zhang, J.; Nie, Y.; Hu, J.; Wang, S.; Zhang, R. A new tetrodotoxin-producing actinomycete, Nocardiopsis dassonvillei, isolated from the ovaries of puffer fish Fugu rubripes. Toxicon 2005, 45, 851–859. [Google Scholar] [CrossRef]
  93. Noguchi, T.; Arakawa, O. Tetrodotoxin—Distribution and accumulation in aquatic organisms and cases of human intoxication. Mar. Drugs 2008, 6, 220–242. [Google Scholar] [CrossRef]
  94. Hwang, D.F.; Arakawa, O.; Saito, T.; Noguchi, T.; Simidu, U.; Tsukamoto, K.; Shida, Y.; Hashimoto, K. Tetrodotoxin-producing bacteria from the blue-ringed octopus Octopus maculosus. Mar. Biol. Heidelb. Ger. 1989, 100, 327–332. [Google Scholar] [CrossRef]
  95. Do, H.K.; Hamasaki, K.; Ohwada, K.; Simidu, U.; Noguchi, T.; Shida, Y.; Kogure, K. Presence of tetrodotoxin and tetrodotoxin-producing bacteria in freshwater sediments. Appl. Environ. Microbiol. 1993, 59, 3934–3937. [Google Scholar]
  96. Hasan, S.; Nikkon, F.; Pervin, F.; Rahman, M.M.; Khatun, S.; Hossain, T.; Khan, A.; Sarker, S.K.; Mosaddik, A.; Absar, N. Biochemical and histopathological effects of tetrodotoxin isolated from puffer fish Tetraodon patoca available in Bangladesh. Res. J. Med. Med. Sci. 2008, 3, 177–181. [Google Scholar]
  97. Zimmer, T. Effects of tetrodotoxin on the mammalian cardiovascular system. Mar. Drugs 2010, 8, 741–762. [Google Scholar] [CrossRef]
  98. Denac, H.; Mevissen, M.; Scholtysik, G. Structure, function and pharmacology of voltage-gated sodium channels. Pharmacol. 2000, 362, 453–479. [Google Scholar]
  99. Danovaro, R.; Fonda Umani, S.; Pusceddu, A. Climate change and the potential spreading of marine mucilage and microbial pathogens in the mediterranean sea. PLoS ONE 2009, 4, 1–8. [Google Scholar]
  100. O’Leary, M.A.; Schneider, J.J.; Isbister, G.K. Use of high performance liquid chromatography to measure tetrodotoxin in serum and urine of poisoned patients. Toxicon 2004, 44, 549–553. [Google Scholar] [CrossRef]
  101. Isbister, G.K.; Son, J.; Wang, F.; Maclean, C.J.; Lin, C.S.; Ujma, J.; Balit, C.R.; Smith, B.; Milder, D.G.; Kiernan, M.C. Puffer fish poisoning: A potentially life-threatening condition. Med. J. Aust. 2002, 177, 650–653. [Google Scholar]
  102. Chowdhury, F.R.; Nazmul Ahasan, H.A.; Mamunur Rashid, A.K.; Al Mamun, A.; Khaliduzzaman, S.M. Tetrodotoxin poisoning: A clinical analysis, role of neostigmine and short-term outcome of 53 cases. Singapore Med. J. 2007, 48, 830–833. [Google Scholar]
  103. Arakawa, O.; Hwang, D.-F.; Taniyama, S.; Takatani, T. Toxins of Pufferfish that Cause Human Intoxications. In Coastal Environmental and Ecosystem Issues of the East China Sea; Ishimatsu, A., Lie, H.-J., Eds.; Terrapub and Nagasaki University: Tokyo, Japan, 2010; pp. 227–244. [Google Scholar]
  104. Cho, H.E.; Ahn, S.Y.; Son, I.S.; In, S.; Hong, R.S.; Kim, D.W.; Woo, S.H.; Moon, D.C.; Kim, S. Determination and validation of tetrodotoxin in human whole blood using hydrophilic interaction liquid chromatography-tandem mass spectroscopy and its application. Forensic Sci. Int. 2012, 217, 76–80. [Google Scholar] [CrossRef]
  105. Bentur, Y.; Ashkar, J.; Lurie, Y.; Levy, Y.; Azzam, Z.S.; Litmanovich, M.; Golik, M.; Gurevych, B.; Golani, D.; Eisenman, A. Lessepsian migration and tetrodotoxin poisoning due to Lagocephalus sceleratus in the eastern Mediterranean. Toxicon 2008, 52, 964–968. [Google Scholar] [CrossRef]
  106. Tsai, Y.H.; Hwang, D.F.; Cheng, C.A.; Hwang, C.C.; Deng, J.F. Determination of tetrodotoxin in human urine and blood using C18 cartridge column, ultrafiltration and LC-MS. J. Chromatogr. B 2006, 832, 75–80. [Google Scholar] [CrossRef]
  107. Hwang, D.-F.; Noguchi, T.; Steve, L.T. Tetrodotoxin Poisoning. In Advances in Food and Nutrition Research; Elsevier: Lincoln, NE, USA, 2007; Volume 52, pp. 141–236. [Google Scholar]
  108. Cohen, N.J.; Deeds, J.R.; Wong, E.S.; Hanner, R.H.; Yancy, H.F.; White, K.D.; Thompson, T.M.; Wahl, M.; Pham, T.D.; Guichard, F.M.; et al. Public health response to puffer fish (tetrodotoxin) poisoning from mislabeled product. J. Food Prot. 2009, 72, 810–817. [Google Scholar]
  109. Centres for disease control and prevention. Morbidity and Mortality Weekly Report; Centres for disease control and prevention: Atlanta, GA, USA, 17 May 1996.
  110. Sims, J.K.; Ostman, D.C. Pufferfish poisoning: Emergency diagnosis and management of mild human tetrodotoxication. Ann. Emerg. Med. 1986, 15, 1094–1098. [Google Scholar] [CrossRef]
  111. Chau, R.; Kalaitzis, J.A.; Neilan, B.A. On the origins and biosynthesis of tetrodotoxin. Aquat. Toxicol. 2011, 104, 61–72. [Google Scholar] [CrossRef]
  112. Yotsu-Yamashita, M.; Sugimoto, A.; Takai, A.; Yasumoto, T. Effects of specific modifications of several hydroxyls of tetrodotoxin on its affinity to rat brain membrane. J. Pharmacol. Exp. Ther. 1999, 289, 1688–1696. [Google Scholar]
  113. Kishi, Y.; Aratani, M.; Fukuyama, T.; Nakatsubo, F.; Goto, T.; Inoue, S.; Tanino, H.; Sugiura, S.; Kakoi, H. Synthetic studies on tetrodotoxin and related compounds. III. Stereospecific synthesis of an equivalent of acetylated tetrodamine. J. Am. Chem. Soc. 1972, 94, 9217–9219. [Google Scholar] [CrossRef]
  114. Nishikawa, T.; Urabe, D.; Yoshida, K.; Iwabuchi, T.; Asai, M.; Isobe, M. Stereocontrolled synthesis of 8,11-dideoxytetrodotoxin, unnatural analogue of puffer fish toxin. Org. Lett. 2002, 4, 2679–2682. [Google Scholar] [CrossRef]
  115. Hinman, A.; Du Bois, J. A stereoselective synthesis of (-)-tetrodotoxin. J. Am. Chem. Soc. 2003, 125, 11510–11511. [Google Scholar] [CrossRef]
  116. Nishikawa, T.; Urabe, D.; Yoshida, K.; Iwabuchi, T.; Asai, M.; Isobe, M. Total syntheses of 11-deoxytetrodotoxin and 8,11-dideoxytetrodotoxin. Pure Appl. Chem. 2003, 75, 251–257. [Google Scholar] [CrossRef]
  117. Ohyabu, N.; Nishikawa, T.; Isobe, M. First asymmetric total synthesis of tetrodotoxin. J. Am. Chem. Soc. 2003, 125, 8798–8805. [Google Scholar] [CrossRef]
  118. Nishikawa, T.; Urabe, D.; Isobe, M. An efficient total synthesis of optically active tetrodotoxin. Angew. Chem. Int. Ed. 2004, 43, 4782–4785. [Google Scholar] [CrossRef]
  119. Sato, K.; Akai, S.; Shoji, H.; Sugita, N.; Yoshida, S.; Nagai, Y.; Suzuki, K.; Nakamura, Y.; Kajihara, Y.; Funabashi, M.; et al. Stereoselective and efficient total synthesis of optically active tetrodotoxin from d-Glucose. J. Org. Chem. 2008, 73, 1234–1242. [Google Scholar] [CrossRef]
  120. Adachi, M.; Imazu, T.; Isobe, M.; Nishikawa, T. An improved synthesis of (-)-5,11-dideoxytetrodotoxin. J. Org. Chem. 2013, 78, 1699–1705. [Google Scholar] [CrossRef]
  121. Raybould, T.J.; Bignami, G.S.; Inouye, L.K.; Simpson, S.B.; Byrnes, J.B.; Grothaus, P.G.; Vann, D.C. A monoclonal antibody-based immunoassay for detecting tetrodotoxin in biological samples. J. Clin. Lab. Anal. 1992, 6, 65–72. [Google Scholar] [CrossRef]
  122. Cheun, B.S.; Loughran, M.; Hayashi, T.; Nagashima, Y.; Watanabe, E. Use of a channel biosensor for the assay of paralytic shellfish toxins. Toxicon 1998, 36, 1371–1381. [Google Scholar] [CrossRef]
  123. Campbell, K.; Barnes, P.; Haughey, S.A.; Higgins, C.; Kawatsu, K.; Vasconcelos, V.; Elliott, C.T. Development and single laboratory validation of an optical biosensor assay for tetrodotoxin detection as a tool to combat emerging risks in European seafood. Anal. Bioanal. Chem. 2013, 405, 7753–7763. [Google Scholar] [CrossRef]
  124. Yotsu-Yamashita, M.; Yamagishi, Y.; Yasumoto, T. 5,6,11-Trideoxytetrodotoxin from the puffer fish, Fugu poecilonotus. Tetrahedron Lett. 1995, 36, 9329–9332. [Google Scholar] [CrossRef]
  125. Jang, J.-H.; Yotsu-Yamashita, M. 6,11-Dideoxytetrodotoxin from the puffer fish, Fugu pardalis. Toxicon 2007, 50, 947–951. [Google Scholar] [CrossRef]
  126. Malik, A.K.; Blasco, C.; Picó, Y. Liquid chromatography-mass spectrometry in food safety. J. Chromatogr. A 2010, 1217, 4018–4040. [Google Scholar] [CrossRef]
  127. Chen, X.-W.; Liu, H.-X.; Jin, Y.-B.; Li, S.-F.; Bi, X.; Chung, S.; Zhang, S.-S.; Jiang, Y.-Y. Separation, identification and quantification of tetrodotoxin and its analogs by LC-MS without calibration of individual analogs. Toxicon 2011, 57, 938–943. [Google Scholar] [CrossRef]
  128. Yotsu-Yamashita, M.; Jang, J.-H.; Cho, Y.; Konoki, K. Optimization of simultaneous analysis of tetrodotoxin, 4-epitetrodotoxin, 4,9-anhydrotetrodotoxin, and 5,6,11-trideoxytetrodotoxin by hydrophilic interaction liquid chromatography-tandem mass spectrometry. Forensic Toxicol. 2011, 29, 61–64. [Google Scholar] [CrossRef]
  129. Kao, C.Y.; Furman, F.A. Pharmacological studies on tetrodotoxin, a potent neurotoxin. J. Pharmacol. Exp. Ther. 1963, 140, 31–40. [Google Scholar]
  130. Xu, Q.-H.; Zhao, X.-N.; Wei, C.-H.; Rong, K.-T. Immunologic protection of anti-tetrodotoxin vaccines against lethal activities of oral tetrodotoxin challenge in mice. Int. Immunopharmacol. 2005, 5, 1213–1224. [Google Scholar] [CrossRef]
  131. Katikou, P.; Georgantelis, D.; Sinouris, N.; Petsi, A.; Fotaras, T. First report on toxicity assessment of the Lessepsian migrant pufferfish Lagocephalus sceleratus (Gmelin, 1789) from European waters (Aegean Sea, Greece). Toxicon 2009, 54, 50–55. [Google Scholar] [CrossRef]
  132. Guzmán, A.; de Henestrosa, A.R.F.; Marín, A.-P.; Ho, A.; Borroto, J.I.G.; Carasa, I.; Pritchard, L. Evaluation of the genotoxic potential of the natural neurotoxin Tetrodotoxin (TTX) in a battery of in vitro and in vivo genotoxicity assays. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2007, 634, 14–24. [Google Scholar] [CrossRef]
  133. Woodward, R.B. The structure of tetrodotoxin. Pure Appl. Chem. 1964, 9, 49–74. [Google Scholar] [CrossRef]
  134. Wang, J.; Araki, T.; Tatsuno, R.; Nina, S.; Ikeda, K.; Hamasaki, M.; Sakakura, Y.; Takatani, T.; Arakawa, O. Transfer profile of intramuscularly administered tetrodotoxin to artificial hybrid specimens of pufferfish, Takifugu rubripes and Takifugu niphobles. Toxicon 2011, 58, 565–569. [Google Scholar] [CrossRef]
  135. Fong, B.M.-W.; Tam, S.; Tsui, S.-H.; Leung, K.S.-Y. Development and validation of a high-throughput double solid phase extraction-liquid chromatography-tandem mass spectrometry method for the determination of tetrodotoxin in human urine and plasma. Talanta 2011, 83, 1030–1036. [Google Scholar] [CrossRef]
  136. Taylor, A.D.; Vaisocherová, H.; Deeds, J.; DeGrasse, S.; Jiang, S. Tetrodotoxin detection by a surface plasmon resonance sensor in pufferfish matrices and urine. J. Sens. 2011, 2011, 1–10. [Google Scholar]
  137. Man, C.N.; Noor, N.M.; Harn, G.L.; Lajis, R.; Mohamad, S. Screening of tetrodotoxin in puffers using gas chromatography-mass spectrometry. J. Chromatogr. A 2011, 1217, 7455–7459. [Google Scholar]
  138. Diener, M.; Christian, B.; Ahmed, M.; Luckas, B. Determination of tetrodotoxin and its analogs in the puffer fish Takifugu oblongus from Bangladesh by hydrophilic interaction chromatography and mass-spectrometric detection. Anal. Bioanal. Chem. 2007, 389, 1997–2002. [Google Scholar] [CrossRef]
  139. Tsai, Y.-H.; Ho, P.-H.; Hwang, C.-C.; Hwang, P.-A.; Cheng, C.-A.; Hwang, D.-F. Tetrodotoxin in several species of xanthid crabs in southern Taiwan. Food Chem. 2006, 95, 205–212. [Google Scholar] [CrossRef]
  140. Asakawa, M.; Toyoshima, T.; Ito, K.; Bessho, K.; Yamaguchi, C.; Tsunetsugu, S.; Shida, Y.; Kajihara, H.; Mawatari, S.F.; Noguchi, T.; et al. Paralytic toxicity in the ribbon worm Cephalothrix species (Nemertea) in Hiroshima Bay, Hiroshima Prefecture, Japan and the isolation of tetrodotoxin as a main component of its toxins. Toxicon 2003, 41, 747–753. [Google Scholar] [CrossRef]
  141. Asakawa, M.; Ito, K.; Kajihara, H. Highly toxic ribbon worm Cephalothrix simula containing tetrodotoxin in Hiroshima Bay, Hiroshima Prefecture, Japan. Toxins 2013, 5, 376–395. [Google Scholar] [CrossRef]
  142. Nakamura, M.; Yasumoto, T. Tetrodotoxin derivatives in puffer fish. Toxicon 1985, 23, 271–276. [Google Scholar] [CrossRef]
  143. Yotsu, M.; Yasumoto, T.; Kim, Y.H.; Naoki, H.; Kao, C.Y. The structure of chiriquitoxin from the Costa Rican frog Atelopus chiriquiensis. Tetrahedron Lett. 1990, 31, 3187–3190. [Google Scholar] [CrossRef]
  144. Hwang, D.F.; Noguchi, T.; Arakawa, O.; Abe, T.; Hashimoto, K. Toxicological studies on several species of puffer in Taiwan. Nippon Suisan Gakkaishi 1988, 54, 2001–2008. [Google Scholar]
  145. Goto, T.; Kishi, Y.; Takahashi, S.; Hirata, Y. Tetrodotoxin. Tetrahedron 1965, 21, 2059–2088. [Google Scholar] [CrossRef]
  146. Pavelka, L.A.; Fuhrman, F.A.; Mosher, H.S. 11- Nortetrodotoxin-6,6-diol and 11-nortetrodotoxin-6-ol. Heterocycles 1982, 17, 225–230. [Google Scholar] [CrossRef]
  147. Endoa, A.; Khora, S.S.; Murata, M.; Naoki, H.; Yasumoto, T. Isolation of 11-nortetrodotoxin-6(R)-OL and other tetrodotoxin derivatives from the puffer Fugu niphobles. Tetrahedron Lett. 1988, 29, 4127–4128. [Google Scholar] [CrossRef]
  148. Yotsu, M.; Hayashi, Y.; Khora, S.S.; Sato, S.i.; Yasumoto, T. Isolation and structural assignment of 11-nortetrodotoxin-6(S)-ol from the puffer Arothron nigropunctatus. Biosci. Biotechnol. Biochem. 1992, 56, 370–371. [Google Scholar] [CrossRef]
  149. Shoji, Y.; Yotsu-Yamashita, M.; Miyazawa, T.; Yasumoto, T. Electrospray ionization mass spectrometry of tetrodotoxin and its analogs: Liquid chromatography/mass spectrometry, tandem mass spectrometry, and liquid chromatography/tandem mass spectrometry. Anal. Biochem. 2001, 290, 10–17. [Google Scholar] [CrossRef]
  150. LATOXAN, France. Available online: http://www.latoxan.com/PHP/00000171.php (accessed on 14 February 2014).
  151. Acros Organics (BVBA), Belgium. Available online: http://wercs.acros.com/wercsdata/document.aspx?prd=ACR25647~~PDF~~MTR~~CLP1~~EN~~2010-12-05%2013:41:44~~Tetrodotoxin,%20citrate%20free (accessed on 14 February 2014).
  152. CMS Chemicals Ltd., UK. Available online: http://www.cms-chemicals.com/cms/website.nsf/%28allcategorycms%29/86C09EFA1D2EE78B802579FA00589A94 (accessed on 14 February 2014).
  153. Hangzhou Dayangchem Co., Zhejiang, China. Available online: http://dayangchem.guidechem.com/pro-show2145147.html (accessed on 14 February 2014).
  154. Acros Organics N.V., Belgium. Available online: https://fscimage.fishersci.com/msds/01139.htm (accessed on 14 February 2014).
  155. Sigma Aldrich Ltd. Available online: http://www.sigmaaldrich.com/catalog/product/sigma/t8024?lang=en&region=IE (accessed on 14 February 2014).
  156. Biotium, USA. Available online: http://www.biotium.com/product/product_info/MSDS/MSDS%2000060.pdf (accessed on 14 February 2014).
  157. Alomone Labs. Available online: http://www.linscottsdirectory.com/products/alomone-labs/tetrodotoxin-citrate-free-14 (accessed on 14 February 2014).
  158. EMD Millipore (Calbiochem®), Germany. Available online: http://www.emdmillipore.com/life-science-research/tetrodotoxin-citrate-free-fugu-sp-/EMD_BIO-584415/p_bAqb.s1LILwAAAEWlWEfVhTm (accessed on 14 February 2014).
  159. Affix Scientific, USA. Available online: http://www.affixscientific.com/product/tetrodotoxin-citrate-fr/ (accessed on 14 February 2014).
  160. Abcam plc, UK. Available online: http://www.abcam.com/Tetrodotoxin-citrate-ab120055.html (accessed on 14 February 2014).
  161. Biorbyt Limited. Available online: http://www.biorbyt.com/tetrodotoxin-citrate-free (accessed on 14 February 2014).
  162. Carbosynth Limited, UK. Available online: http://www.carbosynth.com/carbosynth/website.nsf/%28w-productdisplay%29/86C09EFA1D2EE78B802579FA00589A94 (accessed on 14 February 2014).
  163. Biotium, G.S.-. USA. Available online: http://www.clonagen.com/clonagen/d10e987f-efe7-45f3-b1a9-2f9d3cb88753/tetrodotoxin_citrate_free_product.aspx (accessed on 14 February 2014).
  164. Yotsu-Yamashita, M.; Goto, A.; Nakagawa, T. Identification of 4-S-Cysteinyltetrodotoxin from the liver of the puffer fish, Fufu pardalis and formation of thiol adducts of tetrodotoxin from 4,9-anhydro-tetrodotoxin. Chem. Res. Toxicol. 2005, 18, 865–871. [Google Scholar] [CrossRef]
  165. Khora, S.S.; Yasumoto, T. Isolation of 11-oxotetrodotoxin from the puffer, Arothron nigropunctatus. Tetrahedron Lett. 1989, 30, 4393–4394. [Google Scholar] [CrossRef]
  166. Kono, M.; Matsui, T.; Furukawa, K.; Takase, T.; Yamamori, K.; Kaneda, H.; Aoki, D.; Jang, J.-H.; Yotsu-Yamashita, M. Examination of transformation among tetrodotoxin and its analogs in the living cultured juvenile puffer fish, kusafugu, Fugu niphobles by intramuscular administration. Toxicon 2008, 52, 714–720. [Google Scholar] [CrossRef]
  167. Nakashima, K.; Arakawa, O.; Taniyama, S.; Nonaka, M.; Takatani, T.; Yamamori, K.; Fuchi, Y.; Noguchi, T. Occurrence of saxitoxins as a major toxin in the ovary of a marine puffer Arothron firmamentum. Toxicon 2004, 43, 207–212. [Google Scholar] [CrossRef]
  168. Vázquez, N.; Yotsu-Yamashita, M.; Sierra-Beltrán, A.P.; Yasumoto, T.; Ochoa, J.L. Toxicities and distribution of tetrodotoxin in the tissues of puffer fish found in the coast of the Baja California Peninsula, Mexico. Toxicon 2000, 38, 729–734. [Google Scholar] [CrossRef]
  169. Katsutoshi, I.; Shinya, O.; Manabu, A.; Kentaro, B.; Shigeto, T.; Yasuo, S.; Susumu, O. Detection of tetrodotoxin (TTX) from two copepods infecting the grass puffer Takifugu niphobles: TTX attracting the parasites? Toxicon 2006, 48, 620–626. [Google Scholar] [CrossRef]
  170. Asakawa, M.; Gomez-Delan, G.; Tsuruda, S.; Shimomura, M.; Shida, Y.; Taniyama, S.; Barte-Quilantang, M.; Shindo, J. Toxicity assessment of the Xanthid Crab Demania cultripes from Cebu Island, Philippines. J. Toxicol. 2010, 2010, 1–7. [Google Scholar]
  171. Ho, P.-H.; Tsai, Y.-H.; Hwang, C.-C.; Hwang, P.-A.; Hwang, J.-H.; Hwang, D.-F. Paralytic toxins in four species of coral reef crabs from Kenting National Park in southern Taiwan. Food Control 2006, 17, 439–445. [Google Scholar] [CrossRef]
  172. Kanchanapongkul, J. Tetrodotoxin poisoning following ingestion of the toxic eggs of the horseshoe crab Carcinoscorpius rotundicauda, a case series from 1994 through 2006. Southeast Asian J. Trop. Med. Public Health 2008, 39, 303–306. [Google Scholar]
  173. Yasumoto, T.; Yotsu, M.; Endo, A.; Murata, M.; Naoki, H. lnterspecies distribution and biogenetic origin of tetrodotoxin and its derivatives. Pure Appl. Chem. 1989, 61, 505–508. [Google Scholar] [CrossRef]
  174. Narita, H.; Matsubara, S.; Miwa, N.; Akahane, S.; Murakami, M.; Goto, T.; Nara, M.; Noguchi, T.; Saito, T.; Shida, Y. Vibrio alginolyticus, a TTX-producing bacterium isolated from the starfish Astropecten polyacanthus. Nippon Suisan Gakk. 1987, 53, 617–621. [Google Scholar] [CrossRef]
  175. Nakagawa, T.; Jang, J.; Yotsu-Yamashita, M. Hydrophilic interaction liquid chromatography-electrospray ionization mass spectrometry of tetrodotoxin and its analogs. Anal. Biochem. 2006, 352, 142–144. [Google Scholar] [CrossRef]
  176. Yotsu, M.; Endo, A.; Yasumoto, T. An improved tetrodotoxin analyzer. Agric. Biol. Chem. 1989, 53, 893–895. [Google Scholar] [CrossRef]
  177. Umezawa, T.; Shinada, T.; Ohfune, Y. Synthesis of 13C-Labeled 5,6,11-Trideoxytetrodotoxin. Chem. Lett. 2010, 39, 1281–1282. [Google Scholar] [CrossRef]
  178. Williams, B.L. Behavioral and chemical ecology of marine organisms with respect to tetrodotoxin. Mar. Drugs 2010, 8, 381–398. [Google Scholar] [CrossRef]
  179. Hanifin, C.T. The chemical and evolutionary ecology of tetrodotoxin (TTX) toxicity in terrestrial vertebrates. Mar. Drugs 2010, 8, 577–593. [Google Scholar] [CrossRef]
  180. Childress, J.J.; Fisher, C.R.; Brooks, J.M.; Kennicutt, M.C.; Bidigare, R.; Anderson, A.E. A methanotrophic marine molluscan (Bivalvia, Mytilidae) symbiosis: Mussels fueled by gas. Science 1986, 233, 1306–1308. [Google Scholar]
  181. Ott, J.A.; Bright, M.; Schiemer, F. The ecology of a novel symbiosis between a marine peritrich ciliate and chemoautotrophic bacteria. Mar. Ecol. 1998, 19, 229–243. [Google Scholar] [CrossRef]
  182. Dubilier, N.; Bergin, C.; Lott, C. Symbiotic diversity in marine animals: The art of harnessing chemosynthesis. Nat. Rev. Microbiol. 2008, 6, 725–740. [Google Scholar] [CrossRef]
  183. Matsumura, K. Production of tetrodotoxin in puffer fish embryos. Environ. Toxicol. Pharmacol. 1998, 6, 217–219. [Google Scholar] [CrossRef]
  184. Cheng, C.A.; Lin, S.J.; Hwang, D.F. Paralytic toxins of the gastropod Natica lineata in Pingtung Prefecture. Food Sci. 1996, 26, 845–853. [Google Scholar]
  185. Caldwell, J.P. The evolution of myrmecophagy and its correlates in poison frogs (Family Dendrobatidae). J. Zool. 1996, 240, 75–101. [Google Scholar] [CrossRef]
  186. Hille, B. The permeability of the sodium channel to organic cations in myelinated nerve. J. Gen. Physiol. 1971, 58, 599–619. [Google Scholar] [CrossRef]
  187. Moran, O.; Picollo, A.; Conti, F. Tonic and phasic guanidinium toxin-block of skeletal muscle Na channels expressed in Mammalian cells. Biophys. J. 2003, 84, 2999–3006. [Google Scholar] [CrossRef]
  188. Matsui, T.; Yamamori, K.; Furukawa, K.; Kono, M. Purification and some properties of a tetrodotoxin binding protein from the blood plasma of kusafugu, Takifugu niphobles. Toxicon 2000, 38, 463–468. [Google Scholar] [CrossRef]
  189. Yotsu-Yamashita, M.; Sugimoto, A.; Terakawa, T.; Shoji, Y.; Miyazawa, T.; Yasumoto, T. Purification, characterization, and cDNA cloning of a novel soluble saxitoxin and tetrodotoxin binding protein from plasma of the puffer fish, Fugu pardalis. Eur. J. Biochem. 2001, 268, 5937–5946. [Google Scholar] [CrossRef]
  190. Kaneko, Y.; Matsumoto, G.; Hanyu, Y. TTX resistivity of Na/ channel in newt retinal neuron. Biochem. Biophys. Res. Commun. 1997, 240, 651–656. [Google Scholar] [CrossRef]
  191. Maruta, S.; Yamaoka, K.; Yotsu-Yamashita, M. Two critical residues in p-loop regions of puffer fish Na+ channels on TTX sensitivity. Toxicon 2008, 51, 381–387. [Google Scholar] [CrossRef]
  192. Nagashima, Y.; Yamamoto, K.; Shimakura, K.; Shiomi, K. A tetrodotoxin-binding protein in the hemolymph of shore crab Hemigrapsus sanguineus: Purification and properties. Toxicon 2002, 40, 753–760. [Google Scholar] [CrossRef]
  193. Yotsu-Yamashita, M.; Shoji, Y.; Terakawa, T.; Yamada, S.; Miyazawa, T.; Yasumoto, T. Mutual binding inhibition of tetrodotoxin and saxitoxin to their binding protein from the plasma of the puffer fish, Fugu pardalis. Biosci. Biotechnol. Biochem. 2002, 66, 2520–2524. [Google Scholar] [CrossRef]
  194. Hwang, P.-A.; Tsai, Y.-H.; Lin, H.-P.; Hwang, D.-F. Tetrodotoxin-binding proteins isolated from five species of toxic gastropods. Food Chem. 2007, 103, 1153–1158. [Google Scholar] [CrossRef]
  195. Fukuda, A.; Tani, A. Records of puffer poisonings report 3. Nippon Igaku Oyobi Kenko Hoken 1941, 3528, 7–13. [Google Scholar]
  196. Leung, K.S.-Y.; Fong, B.M.-W.; Tsoi, Y.-K. Analytical challenges: Determination of tetrodotoxin in human urine and plasma by LC-MS/MS. Mar. Drugs 2011, 9, 2291–2303. [Google Scholar] [CrossRef]
  197. ExPASy, S.B.R.P. Sodium/potassium-exchanging ATPase. Available online: http://enzyme.expasy.org/EC/3.6.3.9 (accessed on 14 February 2014).
  198. Meyer, T.W.; Hostetter, T.H. Uremia. N. Engl. J. Med. 2007, 357, 1316–1325. [Google Scholar] [CrossRef]
  199. Nakashima, R.; Nakata, Y.; Kameoka, M.; Hayashi, N.; Watanabe, K.; Yagi, K. Case of tetrodotoxin intoxication in a uremic patient. Chudoku Kenkyu 2007, 20, 141–145. [Google Scholar]
  200. Chew, S.K.; Chew, L.S.; Wang, K.W.; Mah, P.K.; Tan, B.Y. Antocholinesterase drugs in the treatement of tetrodotoxin poisoning. Lancet 1984, 324. [Google Scholar] [CrossRef]
  201. Clark, R.F.; Williams, S.R.; Nordt, S.P.; Manoguerra, A.S. A review of selected sea-food poisoning. Undersea Hyperb. Med. 1999, 26, 175–184. [Google Scholar]
  202. Vale, J.A. Position statement: Gastric lavage. American academy of clinical toxicology; European association of poisons centres and clinical toxicologists. J. Toxicol. Clin. Toxicol. 1997, 35, 711–719. [Google Scholar] [CrossRef]
  203. Kaufman, B.; Wright, D.C.; Ballou, W.R.; Monheit, D. Protection against tetrodotoxin and saxitoxin intoxication by a cross-protective rabbit anti-tetrodotoxin antiserum. Toxicon 1991, 29, 581–587. [Google Scholar] [CrossRef]
  204. Rivera, V.R.; Poli, M.A.; Bignami, G.S. Prophylaxis and treatment with a monoclonal antibody of tetrodotoxin poisoning in mice. Toxicon 1995, 33, 1231–1237. [Google Scholar] [CrossRef]
  205. Joshi, S.K.; Joseph, P.M.; Hernandez, G.; Baker, S.; Shieh, C.-C.; Neelands, T.; Zhang, X.-F.; Niforatos, W.; Kage, K.; Han, P.; et al. Involvement of the TTX-resistant sodium channel Nav 1.8 in inflammatory and neuropathic, but not post-operative, pain states. Pain 2006, 123, 75–82. [Google Scholar] [CrossRef]
  206. Marcil, J.; Walczak, J.-S.; Guindon, J.; Ngoc, A.H.; Lu, S.; Beaulieu, P. Antinociceptive effects of tetrodotoxin (TTX) in rodents. Br. J. Anaesth. 2006, 96, 761–768. [Google Scholar] [CrossRef]
  207. Hagen, N.A.; du Souich, P.; Lapointe, B.; Ong-Lam, M.; Dubuc, B.; Walde, D.; Love, R.; Ngoc, A.H. Tetrodotoxin for moderate to severe cancer pain: A randomized, double blind, parallel design multicenter study. J. Pain Symptom Manag. 2008, 35, 420–429. [Google Scholar] [CrossRef]
  208. Shi, J.; Liu, T.T.; Wang, X.; Epstein, D.H.; Zhao, L.Y.; Zhang, X.L.; Lu, L. Tetrodotoxin reduces cue-induced drug craving and anxiety in abstinent heroin addicts. Pharmacol. Biochem. Behav. 2009, 92, 603–607. [Google Scholar] [CrossRef]
  209. Por, F.D.E. Lessepsian Migration, the Influx of Red Sea Biota into the Mediterranean by Way of the Suez Canal; Springer-Verlag: Berlin, Germany, 1978. [Google Scholar]
  210. Akyol, O.; Ünal, V.; Ceyhan, T.; Bilecenoglu, M. First confirmed record of Lagocephalus sceleratus (Gmelin, 1789) in the Mediterranean Sea. J. Fish Biol. 2005, 66, 1183–1186. [Google Scholar] [CrossRef]
  211. Golani, D.; Levy, Y. New records and rare occurrences of fish species from the Mediterranean coast of Israel. Zool. Middle East 2005, 36, 27–32. [Google Scholar] [CrossRef]
  212. Corsini, M.; Margies, P.; Kondilatos, G.; Economidis, P.S. Three new exotic fish records from the SE Aegean Greek waters. Sci. Mar. 2006, 70, 319–323. [Google Scholar]
  213. Peristeraki, P.; Lazarakis, G.; Tserpes, G. First Results on the maturity of the Lessepsian migrant Lagocephalus sceleratus (gmelin 1789) in the eastern meditettanean sea. Rapp. Comm. int. Mer Médit 2010, 39, 628. [Google Scholar]
  214. Jribi, I.; Bradai, M.N. First record of the Lessepsian migrant species Lagocephalus sceleratus (Gmelin, 1789) (Actinopterygii: Tetraodontidae) in the Central Mediterranean. BioInvasions Rec. 2012, 1, 49–52. [Google Scholar] [CrossRef]
  215. Milazzo, M.; Azzurro, E.; Badalamenti, F. On the occurrence of the silverstripe blaasop Lagocephalus sceleratus (Gmelin, 1789) along the Libyan coast. BioInvasions Rec. 2012, 1, 125–127. [Google Scholar] [CrossRef] [Green Version]
  216. Nader, M.R.; Indary, S.; Boustany, L.E. The Puffer Fish Lagocephalus Sceleratus (Gmelin, 1789) in the Eastern Mediterranean; EastMed Technical Documents 2012, GCP/INT/041/EC–GRE–ITA; FAO: Rome, Italy, 2012. [Google Scholar]
  217. Mosher, H.S.; Fuhrman, F.A.; Buchwald, H.D.; Fischer, H.G. Tarichatoxin-tetrodotoxin: A potent neurotoxin. Science 1964, 144, 1100–1110. [Google Scholar]
  218. Nagashima, Y.; Tanaka, N.; Shimakura, K.; Shiomi, K.; Shida, Y. Occurrence of tetrodotoxin-related substances in the nontoxic puffer Takifugu xanthopterus. Toxicon 2001, 39, 415–418. [Google Scholar] [CrossRef]
  219. Simon, K.D.; Mazlan, A.G.; Usup, G. Toxicity of puffer fishes (Lagocephalus wheeleri Abe, tabeta and kitahama, 1984 and Lagocephalus sceleratus Gmelin, 1789) from the east coast waters of peninsular Malaysia. J. Biol. Sci. 2009, 9, 482–487. [Google Scholar] [CrossRef]
  220. Noguchi, T.; Hashimoto, Y. Isolation of tetrodotoxin from a goby Gobius criniger. Toxicon 1973, 11, 305–307. [Google Scholar] [CrossRef]
  221. Thuesen, E.V.; Kogure, K.; Hashimoto, K.; Nemoto, T. Poison arrowworms: A tetrodotoxin venom in the marine phylum Chaetognatha. J. Exp. Mar. Biol. Ecol. 1988, 116, 249–256. [Google Scholar] [CrossRef]
  222. Ngy, L.; Tada, K.; Yu, C.-F.; Takatani, T.; Arakawa, O. Occurrence of paralytic shellfish toxins in Cambodian Mekong pufferfish Tetraodon turgidus: Selective toxin accumulation in the skin. Toxicon 2008, 51, 280–288. [Google Scholar] [CrossRef]
  223. Oliveira, J.S.; Fernandes, S.C.R.; Schwartz, C.A.; Bloch, C., Jr.; Taquita Melo, J.A.; Rodrigues Pires, O., Jr.; Carlos de Freitas, J. Toxicity and toxin identification in Colomesus asellus, an Amazonian (Brazil) freshwater puffer fish. Toxicon 2006, 48, 55–63. [Google Scholar] [CrossRef]
  224. Deeds, J.R.; White, K.D.; Etheridge, S.M.; Landsberg, J.H. Concentrations of saxitoxin and tetrodotoxin in three species of puffers from the Indian River Lagoon, Florida, the location for multiple cases of saxitoxin puffer poisoning from 2002 to 2004. Trans. Am. Fish. Soc. 2008, 137, 1317–1326. [Google Scholar] [CrossRef]
  225. Zaman, L.; Arakawa, O.; Shimosu, A.; Onoue, Y. Occurrence of paralytic shellfish poison in Bangladeshi freshwater puffers. Toxicon 1997, 35, 423–431. [Google Scholar] [CrossRef]
  226. Kungsuwan, A.; Arakawa, O.; Promdet, M.; Onoue, Y. Occurrence of paralytic shellfish poisons in thai freshwater puffers. Toxicon 1997, 35, 1341–1346. [Google Scholar] [CrossRef]
  227. Ito, K.; Asakawa, M.; Beppu, R.; Takayama, H.; Miyazawa, K. PSP-toxicification of the carnivorous gastropod Rapana venosa inhabiting the estuary of Nikoh River, Hiroshima Bay, Hiroshima Prefecture, Japan. Mar. Pollut. Bull. 2004, 48, 1116–1121. [Google Scholar] [CrossRef]
  228. Sato, S.; Kodama, M.; Ogata, T.; Saitanu, K.; Furuya, M.; Hirayama, K.; Kakinuma, K. Saxitoxin as a toxic principle of a freshwater puffer, Tetraodon fangi, in Thailand. Toxicon 1997, 35, 137–140. [Google Scholar] [CrossRef]
  229. Landsberg, J.H.; Hall, S.; Johannessen, J.N.; White, K.D.; Conrad, S.M.; Abbott, J.P.; Flewelling, L.J.; et al. Saxitoxin puffer fish poisoning in the united states, with the first report of Pyrodinium bahamense as the putative toxin source. Environ. Health Perspect. 2006, 114, 1502–1507. [Google Scholar] [CrossRef]
  230. Hille, B. Ionic Channels of Excitable Membranes; Sinauer: Sunderland, MA, USA, 1992. [Google Scholar]
  231. Chen, H.-M.; Lin, C.-H.; Wang, T.M. Effects of 4-aminopyridine on saxitoxin intoxication. Toxicol. Appl. Pharm. 1996, 141, 44–48. [Google Scholar]
  232. Chang, F.-C.T.; Spriggs, D.L.; Benton, B.J.; Keller, S.A.; Capacio, B.R. 4-Aminopyridine reverses saxitoxin (STX)- and tetrodotoxin (TTX)-induced cardiorespiratory depression in chronically instrumented guinea pigs. Fund. Appl. Toxicol. 1997, 38, 75–88. [Google Scholar] [CrossRef]
  233. Tsuda, K.; Ikuma, S.; Kawamura, M.; Tachiikawa, R.; Sakai, K. Tetrodotoxin VII on the structure of tetrodotoxin and its derivatives. Chem. Pharm. Bull. 1964, 12, 1357–1374. [Google Scholar] [CrossRef]
  234. Anraku, K.; Nonaka, K.; Yamaga, T.; Yamamoto, T.; Shin, M.-C.; Wakita, M.; Hamamoto, A.; Akaike, N. Removal of toxin (tetrodotoxin) from puffer ovary by traditional fermentation. Toxins 2013, 5, 193–202. [Google Scholar] [CrossRef]
  235. Doucette, G.J.; Powell, C.L.; Do, E.U.; Byon, C.Y.; Cleves, F.; McClain, S.G. Evaluation of 11-[3H]-tetrodotoxin use in a heterologous receptor binding assay for PSP toxins. Toxicon 2000, 38, 1465–1474. [Google Scholar] [CrossRef]
  236. Kawatsu, K.; Shibata, T.; Hamano, Y. Application of immunoaffinity chromatography for detection of tetrodotoxin from urine samples of poisoned patients. Toxicon 1999, 37, 325–333. [Google Scholar] [CrossRef]
  237. Tanu, M.B.; Mahmud, Y.; Takatani, T.; Kawatsu, K.; Hamano, Y.; Arakawa, O.; Noguchi, T. Localization of tetrodotoxin in the skin of a brackishwater puffer Tetraodon steindachneri on the basis of immunohistological study. Toxicon 2002, 40, 103–106. [Google Scholar] [CrossRef]
  238. Mahmud, Y.; Arakawa, O.; Ichinose, A.; Begum Tanu, M.; Takatani, T.; Tsuruda, K.; Kawatsu, K.; Hamano, Y.; Noguchi, T. Intracellular visualization of tetrodotoxin (TTX) in the skin of a puffer Tetraodon nigroviridis by immunoenzymatic technique. Toxicon 2003, 41, 605–611. [Google Scholar] [CrossRef]
  239. Monaliza, M.; Samsur, M. Toxicity and toxin properties study of puffer fish collected from sabah waters. Health Environ. J. 2011, 2, 14–17. [Google Scholar]
  240. Stokes, A.N.; Williams, B.L.; French, S.S. An improved competitive inhibition enzymatic immunoassay method for tetrodotoxin quantification. Biol. Proced. Online 2012, 14, 3. [Google Scholar] [CrossRef]
  241. Yotsu-Yamashita, M. Chemistry of puffer fish toxin. J. Toxicol. Toxin Rev. 2001, 20, 51–66. [Google Scholar] [CrossRef]
  242. Zaman, L.; Arakawa, O.; Shimosu, A.; Shida, Y.; Onoue, Y. Occurrence of a methyl derivative of saxitoxin in Bangladeshi freshwater puffers. Toxicon 1998, 36, 627–630. [Google Scholar] [CrossRef]
  243. Mohamad, S.; Yasunaga, Y.; Takefumi, S.; Tomohiro, T.; Osamu, A.; Tamao, N. Accumulation and depuration profiles of PSP toxins in the short-necked clam Tapes japonica fed with the toxic dinoflagellate Alexandrium catenella. Toxicon 2006, 48, 323–330. [Google Scholar] [CrossRef]
  244. Akaki, K.; Hatano, K. Determination of tetrodotoxin in puffer-fish tissues, and in serum and urine of intoxicated humans by liquid chromatography with tandem mass spectrometry. Shokuhin Eiseigaku Zasshi 2006, 47, 46–50. [Google Scholar] [CrossRef]
  245. Furey, A.; Moriarty, M.; Bane, V.; Kinsella, B.; Lehane, M. Ion suppression; a critical review on causes, evaluation, prevention and applications. Talanta 2013, 115, 104–122. [Google Scholar] [CrossRef]
  246. Hungerford, J.M. Committee on natural toxins and food allergens, U.S. food and drug administration marine and freshwater toxins. J. AOAC Int. 2006, 89, 248–269. [Google Scholar]
  247. Yakes, B.J.; Deeds, J.; White, K.; DeGrasse, S.L. Evaluation of surface plasmon resonance biosensors for detection of tetrodotoxin in food matrices and comparison to analytical methods. J. Agric. Food Chem. 2010, 59, 839–846. [Google Scholar]
  248. Wildlife, T.T. Rare Scorpion Fish no Fisherman’s Friend. The Times Newspaper—Wildlife, 11 May 2013. [Google Scholar]

Share and Cite

MDPI and ACS Style

Bane, V.; Lehane, M.; Dikshit, M.; O'Riordan, A.; Furey, A. Tetrodotoxin: Chemistry, Toxicity, Source, Distribution and Detection. Toxins 2014, 6, 693-755. https://doi.org/10.3390/toxins6020693

AMA Style

Bane V, Lehane M, Dikshit M, O'Riordan A, Furey A. Tetrodotoxin: Chemistry, Toxicity, Source, Distribution and Detection. Toxins. 2014; 6(2):693-755. https://doi.org/10.3390/toxins6020693

Chicago/Turabian Style

Bane, Vaishali, Mary Lehane, Madhurima Dikshit, Alan O'Riordan, and Ambrose Furey. 2014. "Tetrodotoxin: Chemistry, Toxicity, Source, Distribution and Detection" Toxins 6, no. 2: 693-755. https://doi.org/10.3390/toxins6020693

Article Metrics

Back to TopTop