botryococcus braunii: a renewable source of hydrocarbons and …ychisti/botryo.pdf · 2002. 10....

35
245 0738-8551/02/$.50 © 2002 by CRC Press LLC Critical Reviews in Biotechnology, 22(3):245–279 (2002) Botryococcus braunii: A Renewable Source of Hydrocarbons and Other Chemicals Anirban Banerjee, 1 Rohit Sharma, 1 Yusuf Chisti, 2 and U. C. Banerjee 1* 1 Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar (Mohali) 160062, Punjab, India; 2 Institute of Technology and Engineering, Massey University, Private Bag 11 222 Palmerston North, New Zealand * Corresponding author: Telephone: 91-172-214682, Fax: 91-172-214692, E-mail address: [email protected] TABLE OF CONTENTS I. Introduction .............................................................................................................. 246 II. Races of B. braunii ................................................................................................... 247 III. Biosynthesis and Accumulation of Unsaturated Hydrocarbons ......................... 248 IV. Cell Physiology and the Extracellular Matrix ...................................................... 254 V. Synthesis of Other Biochemicals ............................................................................ 257 A. Exopolysaccharides .............................................................................................. 257 B. Alkanes ................................................................................................................. 264 C. Growth-Promoting Substances ............................................................................. 264 VI. Role of Bacterial Association in Hydrocarbon Production ................................. 264 VII. Bioreactor Culture of B. braunii ............................................................................ 266 A. Culture Media and Conditions ............................................................................. 266 B. Photobioreactors Used for Cultivation ................................................................ 269 VIII. Use of Immobilized Algal Cells .............................................................................. 269 IX. Downstream Processing ........................................................................................... 270 A. Hydrocarbon Recovery ........................................................................................ 270 B. Converting Algal Hydrocarbons and Oil-Rich Biomass to Fuels ....................... 272 X. Cloning of the Hydrocarbon Genes ....................................................................... 272 XI. Concluding Remarks ................................................................................................ 273 References .............................................................................................................................. 274 ABSTRACT: Botryococcus braunii, a green colonial microalga, is an unusually rich renewable source of hydrocarbons and other chemicals. Hydrocarbons can constitute up to 75% of the dry mass of B. braunii. This review details the various facets of biotechnology of B. braunii, including its microbiology and physiology; production of hydrocarbons and other compounds by the alga; methods of culture; downstream recovery and processing of algal hydrocarbons; and cloning of the algal genes

Upload: others

Post on 26-Mar-2021

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

245

0738-8551/02/$.50© 2002 by CRC Press LLC

Critical Reviews in Biotechnology, 22(3):245–279 (2002)

Botryococcus braunii: A Renewable Source ofHydrocarbons and Other Chemicals

Anirban Banerjee,1 Rohit Sharma,1 Yusuf Chisti,2 and U. C. Banerjee1*

1Department of Biotechnology, National Institute of Pharmaceutical Education and Research,Sector 67, S.A.S. Nagar (Mohali) 160062, Punjab, India; 2Institute of Technology and Engineering,Massey University, Private Bag 11 222 Palmerston North, New Zealand

* Corresponding author: Telephone: 91-172-214682, Fax: 91-172-214692, E-mail address: [email protected]

TABLE OF CONTENTS

I. Introduction .............................................................................................................. 246

II. Races of B. braunii ................................................................................................... 247

III. Biosynthesis and Accumulation of Unsaturated Hydrocarbons ......................... 248

IV. Cell Physiology and the Extracellular Matrix ...................................................... 254

V. Synthesis of Other Biochemicals ............................................................................ 257

A. Exopolysaccharides .............................................................................................. 257

B. Alkanes ................................................................................................................. 264

C. Growth-Promoting Substances ............................................................................. 264

VI. Role of Bacterial Association in Hydrocarbon Production ................................. 264

VII. Bioreactor Culture of B. braunii ............................................................................ 266

A. Culture Media and Conditions ............................................................................. 266

B. Photobioreactors Used for Cultivation ................................................................ 269

VIII. Use of Immobilized Algal Cells .............................................................................. 269

IX. Downstream Processing ........................................................................................... 270

A. Hydrocarbon Recovery ........................................................................................ 270

B. Converting Algal Hydrocarbons and Oil-Rich Biomass to Fuels....................... 272

X. Cloning of the Hydrocarbon Genes ....................................................................... 272

XI. Concluding Remarks ................................................................................................ 273

References .............................................................................................................................. 274

ABSTRACT: Botryococcus braunii, a green colonial microalga, is an unusually rich renewable sourceof hydrocarbons and other chemicals. Hydrocarbons can constitute up to 75% of the dry mass ofB. braunii. This review details the various facets of biotechnology of B. braunii, including itsmicrobiology and physiology; production of hydrocarbons and other compounds by the alga; methodsof culture; downstream recovery and processing of algal hydrocarbons; and cloning of the algal genes

Page 2: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

246

into other microorganisms. B. braunii converts simple inorganic compounds and sunlight to potentialhydrocarbon fuels and feedstocks for the chemical industry. Microorganisms such as B. braunii can,in the long run, reduce our dependence on fossil fuels and because of this B. braunii continues to attractmuch attention.

KEY WORDS: Botryococcus braunii, microalgae, algal hydrocarbons, botryococcene, algal culture,photobioreactors.

I. INTRODUCTION

The unicellular photosynthetic microalgaBotryococcus braunii is member of thechlorophyceae (chlorophyta). Sequence analy-sis of the small subunit of r-RNA (16s RNA)and its comparison with the sequences for otheralgae suggests that B. braunii is closely relatedto Characium vaculatum and Dunaliella parva(Sawayama et al., 1995). This colonialmicroalga is widespread in fresh and brackishwaters of all continents (Chisti, 1980). Thecosmopolitan nature of the algae is confirmedby the strains originating in the USA (Wolf etal., 1985a, b), Portugal, Bolivia, France, IvoryCoast, Morocco, Philippines, Thailand, and theWest Indies (Metzger et al., 1985a). Thesegeographical regions belong to various climaticzones, including the continental, temperate,tropical, and alpine zones.

B. braunii is regarded as a potential sourceof renewable fuel because of its ability to pro-duce large amounts of hydrocarbons. Depend-ing on the strain and growth conditions, up to75% of algal dry mass can be hydrocarbons. Thechemical nature of hydrocarbons varies with theproducer strain. Three races of B. braunii havebeen documented, and these can be differenti-ated on the basis of the characteristic hydrocar-bon they produce. The A race produces odd-numbered C25 to C31, n-alkadienes, and trienes.The B race produces triterpenoid hydrocarbonsknown as botryococcenes (CnH2n–10, n = 30–37),apparently of isoprenoid origin (Chisti, 1980).The L race produces lycopadiene, a C40

tetraterpene.Historically, interest in B. braunii arose

because of its geochemical significance. Paleo-

botanical studies suggest that B. braunii is oneof the major sources of hydrocarbon in a vari-ety of oil-rich deposits dating from the Ordovi-cian period to the present (Cane, 1977).Lycopadiene may be the precursor of lycopanefound in some lacustrine sediment. The occur-rence of botryococcane, C34H70, the hydroge-nated derivative of botryococcene, at levels of0.9 and 1.4% in two Sumatran crude oils hasbeen reported (Moldwan and Seifert, 1980).These concentrations are the highest levels everreported for a single complex fossil biologicalmarker in petroleum (Moldwan and Seifert,1980). Botryococcane has been found in a rangeof coastal bitumen (kerogen) formations inAustralia (Mcirdy et al., 1986). B. braunii isthe main source of C27, C29, C31 alkanes foundin the Pula sediment, a picolene oil shale, ofHungary (Lichitfouse et al., 1994). Recently, aC30 botryococcene related compound, 7,11-cyclobotryococca 5, 12, 26-triene was iso-lated in organic rich sediments from lakeCadagno, Switzerland, revealing the widespreadnature of B. braunii in recent and past environ-ments (Behrens et al., 2000).

Few oil shales contain botryococcene, eventhough B. braunii occurred widely during theformation of those shales. This apparent dis-crepancy is explained by degradation ofbotryococcenes in the oxic conditions of theearly diagenesis (Derenne et al., 1988). Spec-troscopic studies (FTIR, GC-MS, etc.) on un-heated materials and pyrolyzates indicate that apredominant fraction of coorongite (75% of thebitumen free fraction), a rubbery deposit, iscomposed of the degradation-resistant biopoly-mers of the outer walls of B. braunii. These cellwall biopolymers are only marginally affected

Page 3: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

247

by the highly oxic conditions of coorongiteformation.

B. braunii has been reported to convert 3%of the solar energy to hydrocarbons (Gudin etal., 1984). Being a photosynthetic organism,the alga can fix atmospheric CO2. Consequently,burning of algal hydrocarbons is not a net con-tributor to CO2 concentration in the atmosphere.The thermal value of biomass-derived hydro-carbons ranges between 30,000 and 42,000 kJ/kg(Held et al., 1985). Converting a 100-MW ther-mal power plant from using coal to using liquidfuel derived from B. braunii can reduce CO2

emissions by 1.5 × 105 tons/yr (Sawayama etal., 1999). Potentially, the CO2 emission valuescould be reduced further by improving the CO2

fixation efficiency of algae. A substantial ef-fort is being devoted to achieving this goal.

This review discusses the microalgaB. braunii; the synthesis of hydrocarbons andother biochemicals by the alga; the culture con-ditions for the production of hydrocarbons; thedownstream recovery and processing of algalhydrocarbons; the cloning into other microor-ganisms of the B. braunii genes responsible forhydrocarbon synthesis.

II. RACES OF B. braunii

B. braunii exists in three different races:A, B, and L. The races can be differentiatedgenerally on the basis of the characteristic hy-drocarbons they produce. Race A produces C25

to C31, odd-numbered n-alkadienes andalkatrienes. These linear olefins can constituteup to 61% of the dry cell mass of the greenactive-state colonies (Gelpi et al., 1970). Twounusual hydrocarbons, C27H51 triene and C27H48

tetraene, were isolated from a B. braunii strainoriginating in Lake Overjuyo, Bolivia.

The B race produces polymethylated un-saturated triterpenes, called botryococcenes(CnH2n–10, n = 30–37). Botryococcenes can ex-ist as isomers with the same number of carbonsbut different structures. In natural populations,botryococcene can constitute from 27 to 86%

of the dry cell mass (Brown and Knights, 1969).The L race produces a single hydrocarbonC40H78, a tetraterpene, known as lycopadiene(Metzger et al., 1990). This hydrocarbon ac-counts for 2 to 8% of the dry biomass (Metzgerand Casadevall, 1987).

The three races can be differentiated on thebasis of certain morphological and physiologi-cal characteristics (Table 1). The cells of the Lrace (8 to 9 µm × 5 µm in size) are relativelysmaller than the cells of races A and B (13 µm× 7–9 µm) (Metzger et al., 1988). The L racealso contains less pyrenoid (Metzger et al.,1988). Another difference among the races isthe colony color in the stationary phase. The Band L race algae turn red-orange and orange-brownish from green color of the active state.The A race alga turns pale yellow from green.This difference is due to the accumulation ofketo-carotenoids (canthaxanthin, echinenone,adonixanthin, etc.) in the stationary phases ofthe B and L races.

The races can also be distinguished on thebasis of the nature of the biopolymers presentin the cell wall. The A and B races contain longaliphatic compounds cross-linked by etherbridges. In contrast, the basic structure of thebiopolymer of L race is based on lycopadienecross-linked by ether bridges. Long chainalkenyl phenol is detected in the A race. Therole of this aliphatic chain is to increase thesolubility of phenolic moiety in the lipidic re-gion, a requirement for protection against bio-logical and chemical oxidative degradation. Thelipids of B and L races consist of terpenoidsthat are more resistant to bacterial attack thanare the aliphatic hydrocarbons (Metzger andCasadevall, 1989). Although biochemicallydifferent, the various races coexist in naturalenvironment. Mixed populations of alkadieneand botryococcene races have been found inAustralian lakes (Wake and Hillen, 1981). Thecharacteristic hydrocarbons produced by racesA, B, and L of B. braunii are noted in Tables2a, 2b, and 2c, respectively. Because of a gen-erally higher hydrocarbon content, races A and

Page 4: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

248

B may be superior to race L for commercialextraction of hydrocarbon fuel.

III. BIOSYNTHESIS ANDACCUMULATION OF UNSATURATEDHYDROCARBONS

B. braunii A race produces nonisoprenoidhydrocarbons alkadienes and trienes. The struc-tural similarity (locations of double bonds),and stereochemistry of alkadienes resemble thatof oleic acid (18:1, cis-ω9). These features andother evidence suggest that oleic acid is thedirect precursor of the n-alkadienes. The intra-cellular concentration of oleic acid remainsrelatively low during rapid production of hy-drocarbons (e.g., in exponential and early lin-ear phases of growth), but when hydrocarbonproduction declines (deceleration phase) theoleic acid concentration rises sharply (Templieret al., 1984). Other factors (e.g., occurrence ofa terminal double bond in the hydrocarbons;inhibition of production by the presence of

dithioerythritol, DET) suggest that the synthe-sis of hydrocarbons (dienes) proceeds via anelongation-decarboxylation route rather than ahead-to-head condensation pathway. In the elon-gation-decarboxylation mechanism, oleic acidacting as a direct precursor is elongated bysuccessive addition of C2 units derived frommalonyl Co-A until the appropriate chainlengths are attained. These very long chainderivatives are then decarboxylated to givehydrocarbons that are released from elonga-tion-decarboxylation complex (Templier et al.,1984).

The elongation-decarboxylation system ofB. braunii is nonspecific and can accept botholeic acid and elaidic acid (18:1, trans-ω9) toproduce both cis and trans dienes. An isomeriza-tion system exists that converts oleic acid to elaidicacid. This isomerization step is slow relative totransformation of elaidic acid to trans alkadienes,and this prevents accumulation of elaidic acid inthe algal strains that produce trans-dienes. Thestrains lacking in trans dienes (Aus strain) areunable to produce elaidic acid from oleic acid via

TABLE 1Distinctive Features of Races of B. braunii

Page 5: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

249

TABLE 2aThe Major Characteristic Hydrocarbons Produced by Race A of B. braunii

Page 6: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

250

TABLE 2bThe Major Characteristic Hydrocarbons Produced by Race B of B. braunii

Page 7: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

251

TABLE 2b (continued)

Page 8: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

252

isomerization (Templier et al., 1991a, b). Synthe-sis of very long chain monoenic cis, trans-ω9

fatty acids also occurs via a non-specific elonga-tion-decarboxylation system. This system needsa large endogenous pool of the correspondingacids, and it is completely different from thenonspecific elongation-decarboxylation systemthat produces hydrocarbons (Templier et al., 1984).Decarboxylation is the last step of hydrocarbonbiosynthesis. Decarboxylation has a high activa-tion energy and needs the presence of some acti-vating group to facilitate the elimination of car-bon dioxide. The required activation energy maybe provided by a β-OH group present in the longchain fatty acyl derivatives (Yong et al., 1986).

The alkatrienes with one terminal and twocis-ω7, cis-ω9 unsaturations are synthesized byan elongation-decarboxylation mechanism,which is selective. This selective mechanismdoes not accept linoleic acid (cis-ω6, cis-ω9,18:2) as a precursor. Growth in the presence oflarge amounts of exogenous linoleic acid doesnot induce the production of correspondingtrienes. Trienes are also unlikely to be formedvia desaturation of corresponding dienes, asconfirmed by feeding the cells with radiola-belled dienes. Oleic acid is also the direct pre-cursor of trienes. The additional double bond isintroduced probably at the early stages of elon-gation-decarboxylation process, either via di-rect desaturation of oleic acid or desaturationof very long chain fatty acyl derivatives pro-duced by elongation of oleic acid (Templier etal., 1991a).

Race B produces isoprenoid compounds withan unusual 1'-3 fusion of isoprene units in thecenter of the chain. These compounds are knownas botryococcenes. Several biosynthetic mecha-nisms for botryococcenes have been proposed.Studies suggest that the C30 botryococcene, the

parent molecule for the higher homologues, isconstructed from two molecules of farnesyldiphosphate, generated from farnesol by theaction of farnesol phosphokinase (Inone et al.,1994a, b). Presqualene diphosphate, derived fromthe farnesyl diphosphate via carbene addition tothe olefenic double bond (Griessman, 1969), isa member of chrysanthemyl species (1'-2-3 fusedcompounds). The cyclopropylcarbinyl cationgenerated by the release of pyrophosphate leadsto 1'-3 (artemisyl), 2-1'-3 (santolinyl), and 1'-1(head-to-head) structures. In vitro solvolysis stud-ies suggest that 98% of the products have aartemisyl carbon skeleton (Poulter et al., 1977).When a single reactive intermediate can lead tomore than one product, the predominance of aspecific isomer suggests the presence of an en-zyme for regulating the regiochemistry of theintermediate.

Botryococcenes belonging to the artemisylstructure family are formed by a rearrangementof cyclopropylcarbinyl precursor, in this casethe C30 cation (R=C11H19), in a reaction termi-nated by transfer of hydride from NADH orNADPH to the allylic cation (Poulter et al.,1977). Squalene is also derived from the sameprecursor via rearrangement and hydride cap-ture but 1'-1 fusion. Botryococcenes are fa-vored over squalenes because of the more fac-ile rupture of 1'-2 bond. A comparison ofbiosynthesis of botryococcenes and squaleneindicates that the relative positions of the C30

substrate and nicotinamide co-factor are simi-lar in the enzyme substrate complex of squalenesynthase and botryococcene synthase (Huanget al., 1988). A logical inference from this isthat the enzymes catalyzing these reactions mayshare some common structural motifs, includ-ing those important for substrate binding andpossibly catalysis.

TABLE 2cHydrocarbon Produced by Race L of B. braunii

Page 9: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

253

Mutational analysis of amino acids in do-mains III, IV, and V of the squalene synthasesuggests that these regions are essential for theenzyme activity. Asp219 and Asp223 of domainIV appear to be responsible for binding of thediphosphate moiety of farnesyl diphosphate(FPP) through magnesium salt bridges. Tyr171

in domain III is essential for the first step of thereaction. An accumulation of presqualenediphosphate (PSPP) in mutants of Phe288 ofdomain V indicates that this domain is involvedin the second step of the reaction and possiblyin the cleavage of the cyclopropane ring toform 1'-1 linkage.

On the other hand, a synthesis ofbotryococcenes requires facile rupture of theC(1') - C(2') cyclopropane bond (Gu et al.,1998). A possible explanation for the appar-ent discrepancy between biomass accumula-tion and squalene synthase activity is that theculture cycle consists of two growth phases.The first phase would be associated with cellproliferation and accumulation of squalenefor sterol and membrane biogenesis and alimited amount of squalene for extracellularbiopolymer assembly. The early induction ofsqualene synthase could be sufficient to ac-count for that need. The second phase ofbiomass accumulation is correlated withmaturation of cells and a dedication of theirbiosynthetic capacities to the accumulationof botryococcenes.

Given the common mechanistic features, itis not unreasonable to think that botryococcenesand squalene might be derived from a squalenesynthase-like reaction (Jarstfer et al., 1996;Zhang and Poulter, 1995) catalyzed by a singleenzyme that subtly modifies to yield either oneof the two products; however, two differentenzymes may be involved (Figure 1). Also, thealternative procedures involving farnesal and3-hydroxy-2, 3-dihydrofarnesal (both derivedfrom farnesol by the action of farnesolhydratase) acting as precursors (aldol conden-sation followed by rearrangement or MichaelAddition) cannot be overruled (Inone et al.,1993).

The C30 botryococcene acts as the precur-sor for higher botryococcenes. Mono methyla-tions occur in most cases on the carbons 3, 7,16, and 20, other positions involved are 18 (for12), 22 (for 15), and 30 (for 16) (Metzger et al.,1985b). S-adenosyl methionine is reported tobe the methylating agent (Wolf et al., 1985b).Pulse chase experiments suggest that methyla-tions occur one at a time in a sequential man-ner, and the reaction occurs both in the cell andin the extracellular matrix (Metzger et al., 1987).In view of the richness of methylation patternsobserved among this family of hydrocarbons, itis quite possible that methylases in bo-tryococcene pathway have low substrate speci-ficities. The alga does not seem to have a largefamily of highly specific methylases. This issupported by the fact that when the cells arecultivated in presence of high levels of me-thionine, large amounts of methylated squalenesare produced. This occurs because nonspecificmethylases convert squalene to methylatedsqualenes, which divert from the normal sterolbiosynthesis and accumulate with other hydro-carbons (Huang and Poulter, 1989). Cyclic iso-mers occur probably because of alkylation andthe resulting cyclohexyl cation rearranging toolefenic structure (Figure 2) (David et al., 1988;Metzger et al., 1985b; Murakami et al., 1988).

Biosynthesis of lycopadienes is assumed toproceed via tail-to-tail combination of two phy-tyl units; however, a lycopersene reductionmechanism cannot be excluded (Metzger andCasadevall, 1987). Raman spectroscopy andelectron microscopy show that hydrocarbonsaccumulate internally in cytoplasmic inclusions(4%) and externally in successive outer walls(95%) (Largeau et al., 1980). Approximately2% of the hydrocarbons occur as free globulesin the culture medium (Largeau et al., 1980).Hydrocarbons in the external pool can be re-covered by short-contact extraction with sol-vents such as hexane. The hydrocarbons of theexternal and internal pools are structurally thesame, but their relative abundance differs inthese pools. Long chain hydrocarbons predomi-nate in the external pool.

Page 10: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

254

For the B race, an excretory process existsfor botryococcenes from the cell (Metzger et al.,1987). Higher botryococcenes (C34, C36, etc.)accumulate in the external pool, whereas theinternal pool consists of the lower homologues(C30, C31, etc.). In addition to the aforementionedexternal and internal pools where the hydrocar-bons are stored irreversibly, a separate meta-bolic pool exists. In the A race, generation ofalkenyl epoxides from the alkadienes of themetabolic pool is a normal occurrence. How-ever, when the cells are incubated with exog-enous alkadienes, increasing amounts of alkenylepoxide and alkadienones are formed because ofan abnormally large increase in the size of themetabolic pool caused by diffusion of exog-enous alkadienes (Templier et al., 1992a).

IV. CELL PHYSIOLOGY AND THEEXTRACELLULAR MATRIX

The cells of B. braunii are embedded in acommunal extracellular matrix (or “cup”),which is impregnated with oils and cellularexudates (Blackburn, 1936). Cells are attachedto each other by a refringent material that some-times links two or more distinct clumps of cell.The wall of each cell possesses an internalfibrillar layer made of polysaccharide and anexternal trilaminar sheath (TLS) (Largeau etal., 1980). After cell division, each cell secretsnew cups within the cup of the mother cell.Thus, the rubbery matrix appears to be com-posed of successive cups saturated with oil.Cells at the end of the exponential phase con-

FIGURE 1. Mechanism of biosynthesis of botryococcenes and squalene by B. braunii.

Page 11: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

255

tain numerous chloroplasts with thylakoids ar-ranged parallel to the cell surface and very fewstarch granules present. The cytoplasm is filledwith lipid globules (Metzger et al., 1985a, b).

During cultivation, B. braunii undergoes acolor change because of an accumulation ofsecondary carotenoids (carotenoids produced inlarge quantity under stress conditions such asnitrogen deficiency and high light intensity) inthe matrix. The presence of carotenoids is morepronounced in races B and L. In the linear stageof growth, both these races produce almost equalamounts of β, β-carotene, echinenone, 3-OHechinenone, canthaxanthin, lutein, violaxanthin,loroxanthin, and neoxanthin. Whereas lutein isthe major carotenoid in the linear phase (22%and 29% of total in B and L races, respectively),canthaxanthin (46%) together with echinenone(20% and 28%) are the dominating carotenoidsin the stationary phase (Grung et al., 1989).Adonixanthin is also present in substantialamount in the L race in stationary phase (Grunget al., 1994). The main component of extracellu-lar carotenoids is a ketocarotenoid, echinenone.The color change is mainly due to accumulationof echinenone in the intracellular matrix and asimultaneous decrease in the amount of intracel-lular pigments (Tonegawa et al., 1998).

Carotenoid biosynthesis follows the samepathway as botryococcenes’ but with a highercarbon number precursor, geranyl-geranyl py-rophosphate (Poulter and Hughes, 1977). Somenewly identified carotenoids, for example,botryoxanthin-A (Okada et al., 1996),botryoxanthin-B, and α-botryoxanthin-A(Okada et al., 1998), braunixanthin 1 and 2(Okada et al., 1997)—isolated from the B racemay contribute to the color of the algal colo-nies. These new carotenoids are structurallyrelated to squalenes.

The outer wall of B. braunii is composed ofa biopolymer (9 to 12% of dry cell mass) thatis highly resistant to nonoxidative chemicaldegradation, especially acetolysis. The biopoly-mer sometimes resembles sporopollenins be-cause of their modes of deposition, resistantnature, and UV fluorescence. Sporopolleninsare a class of biopolymers originating fromoxidative polymerization of carotenoids and/orcarotenoid esters (Goodway et al., 1973).Sporopollenins make outer walls of spores andpollens and also occur in some algal, bacterial,and fungal cell walls. However, IR and 13CNMR studies reveal that the resistant polymerof B. braunii is not derived from carotenoidsand hence cannot be considered to be sporopol-

FIGURE 2. Cyclization of linear isomers by rearrangement in B. braunii. The numbered structures correspond toidentically numbered compounds in Table 2b.

Page 12: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

256

lenin. All the three races of B. braunii containresistant polymer and this polymer, may be thereason for coorongite formation. A similarityexists between races A and B in the nature ofconstituents of the biopolymer.

Long hydrocarbon chains (up to C31), satu-rated and unbranched, probably linked via etherbridges, make up the polymeric networks ofresistant polymers PRB-A (A race) and PRB-B(B race). Hydroxyl groups and ester groupsalso occur in the biopolymer, but they are ef-fectively protected by the three-dimensionalnetwork of hydrocarbon chains. Consequently,the esters are not affected by drastic saponifi-cation and acidic treatments. Both PRB-A andPRB-B exhibit high hydrogen and low nitro-gen, phosphorus, and oxygen content. Com-parative analysis shows both PRB-A and PRB-B are composed of C24 to C30 very long chainfatty acid derivatives (VLCFA) originating fromoleic acid. Feeding experiments and inhibitionwith dithioerythritol (DTE) suggest that char-acteristic hydrocarbons produced by race Aand PRB-A share some common biosyntheticpathway. On the other hand, no biosyntheticrelationship exists between PRB-B andbotryococcenes, in the B race. Thus, lower la-beling of PRB-A occurs when B. braunii (raceA) is grown in externally added oleic acid (la-beled), reflecting diversion of oleic acid to-ward hydrocarbon production (Lareillard et al.,1988).

The presence of SC1058 (a cinnolinyl acidderivative [1-N-benzyl-3-carboxy-4-ketocinnoline]) in growth medium inhibits the syn-thesis of biopolymer in both A and B races.SC1058 affects the ratio of the external andinternal lipid pools. This effect is much morepronounced in case of A race than B race, re-flecting a strong relationship between the hydro-carbons in A race and PRB-A biosynthesis(Templier et al., 1993). It is suggested thatSC1058 inhibits the transformation of VLCFAderivatives to hydrocarbons by decarboxylation.SC1058 also affects the transport of lipids frominternal to external pool (Templier et al., 1992b).The major constituents of the resistant biopoly-

mer (PRB-A and PRB-B) are botryals, alkenylepoxides, alkenyl phenols, epoxy botryals,alkenyl resorcinols, etc., and the condensationproducts between them and alkadienes,alkatrienes, or long chain fatty acids. The mecha-nism of biosynthesis of PRB-A and PRB-B byB. braunii is given in Figure 3. The cell wall-associated hydrocarbons produced by the vari-ous races of the alga are shown in Table 3.

Botryals (1, as explained in Table 3) areeven-numbered C52 to C64 α-branched, α-un-saturated aldehydes produced by aldol conden-sation of even C26 to C32 monounsaturated al-dehydes. Botryals are analogous to mycolicacid (α-branched, β-OH acids) found in Coryne-bacterium (C32–C36), Nocardia (C34–C36) andMycobacterium (C78–C85) species (Metzger andCasadevall, 1989). The trans isomer (–CHOgroup and bulkier groups are trans) predomi-nates in botryals, which is consistent with acid/base catalyzed aldol condensation mechanism.Alkenyls phenols (5), alkenyl resorcinols (3),and alkenyl pyrogallols (4) are synthesized viaa common pathway, involving a tetraketideintermediate (Figure 4). This symmetrical in-termediate undergoes decarboxylation to re-sorcinol derivatives that, depending on posi-tion of the oxidation, produce alkenylpyrogallols and alkenyl phenols (Metzger andPouet, 1995). These phenolic moieties protectthe aliphatic chains in the A race againstdegradation by bacteria and fungi. Epoxidases,which have broad substrate specificities, areresponsible for the formation of epoxides (6).

Botryococcoid ethers (7) constitute asubstantial fraction of the biomass. Here aC27 alkadienyl secondary alcohol is linkedto the –OH bearing carbon via anotherbridge to a C27 alkatrienyl chain (7a) or toa saturated n-C24/C26 alkyl chains (7b)(Metzger and Casadevall, 1991, 1992). Abiosynthetic mechanism for the formationof botryococcoid ether by B. braunii hasbeen proposed (Figure 5). Botryococcoidethers and other ethers on repeated con-densation produce a network of biopoly-mers. This mechanism includes ring open-

Page 13: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

257

ing of a protonated epoxide with specificcleavage of C-9'-O bond (Metzger andCasadevall, 1992). A resistant biopolymer(PRB-L) occurs in the L race also. Thisaccounts for a larger fraction of total bio-mass than PRB-A and PRB-B do in A andB races. PRB-L structure is composed ofsaturated long (up to C30) hydrocarbonchains probably linked via ether bridges,and it is closely related to lycopadiene, thesole hydrocarbon produced by L race(Dernne et al., 1989). The presence ofepoxylycopane (19), lycopanerol-A (20)(Metzger and Aumelas, 1997), lycopanerolB–G (Rager and Metzger, 2000), and ali-phatic polyaldehyde tetraterpene diolpolyacetal (Bertheas et al., 1999) in PRB-L is also reported.

V. SYNTHESIS OF OTHERBIOCHEMICALS

A. Exopolysaccharides

Microbial exopolysaccharides (EPS) areattracting much attention because of their nu-merous applications. Also, in some cases, theproduction of exopolysaccharides is linked withpathogenicity of microorganisms. The commer-cial usefulness of most industrial polysaccha-rides is based on their ability to alter rheologi-cal characteristics of solutions. Polysaccharidesare used extensively in textiles, laundry prod-ucts, adhesives, paper, paint, and foods. Manyalgae can produce exopolysaccharides, but fewmicroalgal polysaccharides are used commer-cially. EPS produced by the alga Chlamydomo-

FIGURE 3. Mechanism for biosynthesis of PRB-A and PRB-B by B. braunii. The numbered structures correspondto identically numbered compounds in Table 3.

Page 14: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

258

TABLE 3Cell Wall-Associated Hydrocarbon Derivatives Produced by Various Races of B. braunii

Page 15: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

259

TABLE 3 (continued)

Page 16: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

260

TABLE 3 (continued)

Page 17: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

261

TABLE 3 (continued)

Page 18: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

262

nas mexicana can be used as a soil conditioner(Koren and Rayburn, 1984) and EPS synthe-sized by Porphyridium curentum is consideredto be commercially important (Anderson andEakin, 1985).

B. braunii is capable of synthesizingexopolysaccharides, as was first reported forthe A race (Casadevall et al., 1985). Cells ofB. braunii possess an internal fibrillar layermade of mucilaginous polysaccharides. Thispolysaccharide slowly dissolves in the culturemedium, causing an increase in viscosity. Theyield of EPS ranges from 250 g·m–3 for A andB races to 1 kg·m–3 for the L race. Galactose isthe major component of the heterogeneouspolysaccharide. Other detectable componentsof the polymer are fucose, arabinose, rham-nose, uronic acids, and unusual sugars such as3-O-methyl fucose, 3-O-methyl rhamnose, and6-O-methyl hexose. In contrast to A and Braces, glucose is detected in significant amountsin the L race polysaccharides (Allard andCasadevall, 1990). B. braunii UC 58, a strainisolated from a lake in Portugal, produces large

amounts of EPS (4.0 to 5.5 kg·m–3) and lowamounts of hydrocarbons (5%) (Fernandes etal., 1989). The polysaccharide production oc-curs both during growth and stationary phases,but higher production rates are seen as thegrowth rate declines. This enhanced produc-tion is associated with the development of ni-trogen limitation in the culture medium. Undernitrogen starvation, the content of photosyn-thetic pigments in the cell decrease, and therate of photosynthesis is reduced. Microalgaeappear to respond to this condition by degrad-ing nitrogen containing macromolecules andaccumulating carbon reserve compounds, suchas polysaccharides and fats.

Higher growth and production of EPS oc-cur when nitrate is the nitrogen source insteadof urea or ammonium salts. The consumptionof urea and ammonium causes a rapid declinein pH, and this explains the relative poor growthand polymer production when using these ni-trogen sources. The broth viscosity begins todecline when urea and ammonium salts are thenitrogen sources and the culture enters the sta-

FIGURE 4. Mechanism of biosynthesis of alkenyl resorcinol derivatives by B. braunii.

Page 19: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

263

tionary phase. This suggests acid and/or enzy-matic hydrolysis of the biopolymer. During thestationary phase, polysaccharide hydrolyzingenzymes can be released by cell lysis. Nitrateat a concentration of 8 mM is apparently theoptimal nitrogen source for obtaining extendedgrowth (Lupi et al., 1994). Cells grown undercontinuous illumination exhibit higher EPSproductivity (both volumetric and specific) thancells grown under cyclic illumination (Lupi etal., 1994). Optimum temperature range for EPSsynthesis is 25 to 30°C, which is coincidentwith optimum temperature range for growth.Below 25°C EPS synthesis declines, althoughgood growth can still be maintained.

The degree of polymerization of thebiopolymer is significantly reduced when thetemperature is outside the optimal range. Dur-ing batch culture, the rheological behavior ofthe medium changes from Newtonian to non-newtonian because of EPS production duringgrowth; however, in long-term culture the rhe-ology reverts to Newtonian because of hydroly-sis of the polymer (Lupi et al., 1991).

The associated bacteria (Auriobacteriumbarkeri, present in the nonaxenic strain ofB. braunii UC58 isolated from Portugal)have no effect on the production or stimula-tion of EPS synthesis (Fernandes et al.,1989). Supposedly, the affinity of certainbacterial species for sulfated glyco-conju-gates exposed on epithelial cells of suscep-tible hosts can be reduced by microalgalexopolysaccharides. Sulfated polysaccha-rides of microalgae can be used in anti-adhesive therapies against bacterial infec-tions both in cold- and warm-bloodedanimals. The adhesion of human pathogenHelicobacter pylori to the HeLaS3 humancell line is inhibited by various algal polysac-charides (Guzman-Murillo and Ascencio,2000). Because it contains uronic acid, theEPS produced can be used to chelate metalions and reduce metal-associated toxicity.Also, EPS is a potential source of fucose, a6-deoxy sugar, which is a high-value sub-strate in chemical synthesis of flavoringagents (Lupi et al., 1991).

FIGURE 5. Mechanism of biosynthesis of botryococcoid ether by B. braunii.

Page 20: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

264

B. Alkanes

Methylated aldehydes are found in the cellsof B. braunii (Metzger et al., 1991). Thesealdehydes are generated from fatty acids viamethylation by S-adenosyl methionine and thenconverted to aldehydes by fatty acyl reductase.This enzyme requires ATP, CoA, and NADHfor its activity. Fatty acyl reductase has beensolubilized from microsomes by 0.1% octylβ-glucoside and purified to homogeneity byBlue A agarose and palmitoyl CoA agaroseaffinity column chromatography. The molecu-lar weight of the enzyme was determined to be35 kDa by SDS-PAGE. The N-terminal part ofthe enzyme is highly homologous with the fattyacyl reductase isolated from bacteria (Wangand Kolattukudy, 1995).

The aldehydes generated from fatty acidsvia methylation are converted to saturated hy-drocarbons (alkanes). The resulting alkane hasone less carbon than the aldehydes. The alde-hyde to alkane conversion via decarboxyla-tion requires decarbonylase activity underanoxic conditions. The decarbonylase enzymeis located in the microsome, and it is inacti-vated by 8-hydroxyquinoline, O-phenan-throline, and metal chelators such as EDTA.Inactivation by metal chelators reflects theinvolvement of metal ions with the activity ofthe enzyme. Plausibly, the metal ion is a partof the porphyrin complex of the decarbonylaseenzyme. The inactivity of the enzyme in thepresence of oxygen is reflected in an absenceof alkanes when B. braunii is cultivated inaerobic conditions.

The decarbonylase of B. braunii alsocan convert carbon monoxide to carbon di-oxide. This conversion of carbon monoxide,not reported in previously examined hydro-carbon synthesizing systems, may be neces-sary for organisms producing large amountsof hydrocarbons. The decarbonylase has aMichaelis-Menten constant (Km) value of65 µM, a vmax of 1.36 nmol·min–1mg–1

and an optimum pH of 7.0 (Dennis andKolattukudy, 1991).

C. Growth-Promoting Substances

Extracts of B. braunii biomass grown un-der certain conditions have been shown to havea growth-promoting effect on roots (Murakami,2000). Growth promotion is achieved at extractconcentrations as low as 50 ppm (Murakami,2000).

VI. ROLE OF BACTERIALASSOCIATION IN HYDROCARBONPRODUCTION

Interactions between bacteria and algae arewell documented. Bacteria can stimulate algalgrowth by releasing substances such as vitaminsand organic chelating agents (Haines and Guillard,1974). Bacteria can produce assimilable nitrogenderivatives and inorganic nutrients for the algalcells (Parker and Bold, 1961). Also, bacteria caninfluence the pH and the redox potential by pro-ducing carbon dioxide (Parker and Bold, 1961).Bacteria can also inhibit algal growth by compet-ing for limiting nutrients, as the nutrient uptakerates of algae are generally lower than those ofbacteria (Saks and Kahn, 1979). Bacteria cansecrete algaecides (Reim et al., 1974), degradealgal polysaccharides, and decompose algal cells(Fallon and Brock, 1979).

The presence of some accompanying mi-croorganisms in B. braunii cultures was onceclaimed to be at least partly responsible for theobserved production of hydrocarbons (Murryand Thomson, 1977). Studies of batch culturesof B. braunii kutzing (nonaxenic strain) revealthat both the total biomass and hydrocarbonproduction at the onset of stationary phase aresubstantially lower when compared with axeniccontrols (Chirac et al., 1985). A lower hydro-carbon productivity in the presence of bacteriais linked with the hydrocarbon-degrading abil-ity of the bacterial species associated with algalculture (especially Pseudomonas and Flavobac-terium sp.) (Chirac et al., 1985). The variousmicrobial flora associated with B. braunii arelisted in Table 4.

Page 21: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

265

Specific association effects have been stud-ied by co-culturing axenic B. braunii (strain-A)with specific microorganisms such as Flavobac-terium aquatile, Corynebacterium aquatile,Azotobacter chroococcum, and Pseudomonasoleovorans (Jones, 1972). These microorgan-isms were selected for various reasons:F. aquatile and C. aquatile occur commonly inaquatic environments and are known to associ-ate with B. braunii, A. chroococcum can fixnitrogen, and P. oleovorans degrades hydro-carbons. When mixed cultures were aeratedwith CO2-enriched air, only F. aquatile causedenhancements in both the algal biomass andhydrocarbon production relative to axenic con-trols. P. oleovorans caused a marked reductionin algal biomass and hydrocarbon production.

Under CO2 limitation, the CO2 produced bybacterial respiration was used by the alga forphotosynthesis. However, the amount of CO2

generated by bacteria was considerably lowerthan the algal demand. The beneficial effects of

CO2 production outweighed the detrimental ef-fects, and all the CO2-limited mixed culturesshowed considerable increase in biomass andhydrocarbon production (Chirac et al., 1985).

Several bacteria exert considerable influ-ence on growth yield and hydrocarbon produc-tion by B. braunii. The precise effects dependon culture conditions and the microbial speciesinvolved. The presence of Bacillus species andCorynebacterium species can stimulate hydro-carbon biosynthesis. In one case, the presenceof Corynebacterium sp. raised the level of hy-drocarbon of the algal biomass from 5.6% to24.2% (Wang and Xie, 1996). The presence ofmicroorganisms not only influences the quan-tity of hydrocarbons but also their relative abun-dance. Some microorganisms may alter therelative rates of synthesis of major hydrocar-bons by B. braunii. However, the associationwith microorganisms is not essential for thealga to produce large quantities of hydrocar-bons.

TABLE 4Various Microbial Flora Associated with B. braunii

Page 22: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

266

Not all bacterial species can readily associ-ate with B. braunii. The inability to form stableassociation (revealed by a low population ofassociated species) can have several causes,including unsuitable culture conditions, inabil-ity of organic matter released by B. braunii toact as a good nutrient for growth, and antago-nistic effects of B. braunii (Swale, 1968). Inmany cases, stable association will provide alarge increase in algal biomass and hydrocar-bon production, a reduced contamination byother microorganisms, and an improved supplyof CO2.

VII. BIOREACTOR CULTUREB. braunii

A. Culture Media and Conditions

Much work has been done on establishingthe optimal nutritional requirements and cultureconditions for producing B. braunii hydrocar-bons. The production of hydrocarbons inB. braunii appears to be growth associated, irre-spective of the specific culture conditions andthe nutrients used. Apparently, the observed slowgrowth rate of the alga is not a consequence ofa limited supply of nutrients but the result of thesubstantial commitment by the cells to produc-ing energetically expensive hydrocarbons.

Like other microalgae, B. braunii culturerequires water, light, CO2, and inorganic nutri-ents. Culture productivity is affected by factorssuch as pH, pCO2, irradiance, salinity, and tem-perature. A modified Chu-13 medium is oftenused to culture B. braunii. This medium, atfour-fold normal strength, has the followingcomposition (kg·m–3): KNO3 (0.2), K2HPO4

(0.04), MgSO4·7H2O (0.1), CaCl2·6H2O (0.08),ferric citrate (0.01), citric acid (0.1), boron (0.5ppm), manganese (0.5 ppm), copper (0.02 ppm),cobalt (0.02 ppm), and molybdenum (0.02 ppm).The pH is adjusted to 7.5 before sterilization(Largeau et al., 1980).

B. braunii cells grown in a luminostat cul-ture in modified Prat medium attained a biom-

ass concentration of 3.9 kg·m–3 and had a rela-tively short generation time of 3 to 4 days. Thespecific growth rate (µ) during exponentialphase was 0.235 d–1. As the culture aged, itaccumulated carbohydrates and lipids but lostsome nitrogen-containing substances (Volovaet al., 1998).

Photosynthetic cultures of B. braunii requireCO2. Cultures aerated with 0.3% CO2-enrichedair have a much shorter mass doubling time (40 h)compared with 6 days for cultures supplied withambient air. CO2 enrichment favors the forma-tion of lower botryococcenes (C30–C32), whereascultures sparged with ambient air accumulatehigher botryococcenes (C33–C34) (Wolf et al.,1985a). Apparently, methylation steps leadingfrom C30 to C31 and C32 are faster in CO2-en-riched cultures than steps leading to C33, C34 andhigher homologues. Although autotrophic, B.braunii utilizes exogenous carbon sources forimproved growth and hydrocarbon production.Various carbon sources, including C1–C6 com-pounds and disaccharides (lactose, sucrose), havebeen screened in attempts to decrease the massdoubling time of the alga from ≥1 week to lessthan 2 to 3 days (Weetal, 1985).

Although a deficiency of nitrogen favorslipid accumulation (Ben-Amotz et al., 1985),nitrogen is required for growth. Studies withnitrogen supplied as NO3

–, NO2–, and NH3 re-

veal that the primary factor regulating nitrogenmetabolism in B. braunii is the nitrate uptakesystem. Nitrogen is generally supplied as ni-trate salts. An initial NO3

– concentration of≥0.2 kg·m–3 favors hydrocarbon production(Casadevall et al., 1983). With 1 kg·m–3 KNO3,the hydrocarbon concentration after 30 dayswas 4.8 kg·m–3. About the same amount ofhydrocarbon (4.5 kg·m–3) was obtained if theinitial concentration of KNO3 was 3 kg·m–3.This is because at high concentrations nitrateinterferes with hydrocarbon production(Brenckman et al., 1989).

When the cells are exposed to 5 mM NH4+ for

24 h, the nitrate reductase enzyme becomes inac-tive. The use of NH4

+ as the nitrogen sourcecauses the culture pH to decline to less than pH 4,

Page 23: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

267

and this is damaging to cells. This NH4+-related

toxicity manifests itself in the late exponentialphase of growth, and the damage it causes isirreversible (Lupi et al., 1994). Photosynthesisand hydrocarbon production are drastically di-minished when B. braunii (B race) cells areexposed to NH3. Total CO2 fixation declines byabout 60% and oxygen evolution reduces bymore than 50%. However, there is a pronouncedincrease in the synthesis of alanine, glutamine,and other amino acids, especially 5-aminolevulinicacid, a precursor of chlorophyll. This indicates adiversion of acetyl Co-A from hydrocarbon syn-thesis pathway to amino acid synthesis pathway(Ohmori et al., 1984).

Phosphorus is required for the growth ofB. braunii, and it is usually supplied in theform of K2HPO4. Active growth persists aftercomplete exhaustion of phosphate in the me-dium. In fact, phosphate levels can decline tobelow detection (0.5 g·m–3) early in the expo-nential phase (Casadevall et al., 1985). Manyalgae can rapidly absorb phosphate in amountsexceeding the requirement of the cell. This excessphosphate is stored as intracellular granules. Thecells utilize on this phosphate reserve when theextracellular phosphate supply has run out.

In the final stages of culture, a large amountof phosphate can be released in the medium asthe cells lyse. An increase in the initial amountof phosphate in the medium, beyond the levelalready present in the concentrated Chu-13medium, does not affect growth, the nature ofhydrocarbons, and their relative abundance.However, a noticeable increase in the amountof hydrocarbon produced has been reported inthe presence of excess phosphate. This arisesbecause of changes in the N:P ratio in themedium, and N:P ratio is known to influencethe lipid content of various algae (Casadevallet al., 1985).

Studies have attempted to identify the op-timal irradiance levels for supporting growthand hydrocarbon production. A high intensityof light increases the carotenoid-to-chlorophyllratio, and this affects the color of algal colonies(Wolf et al., 1985a). Carbohydrate concentra-

tion, cellular nitrogen, and phosphorus contentof B. braunii UTEX 572 are decreased by ex-tended exposure to a light intensity of 25 to 72µE·m–2s–1 (Oh et al., 1997).

Under intense illumination (10 klx, or ∼140µE·m–2s–1) algal cells that had been adapted toa high irradiance during preculture could attaina higher biomass concentration (7 kg·m–3) andhydrocarbon content (50% of dry weight) com-pared with cells that had been adapted to low-level irradiance (3 klx, or ∼42 µE·m–2s–1)(Kojima and Zhang, 1999). The lower produc-tivity of dark-adapted culture was because itwas highly susceptible to photoinhibition.Photoinhibition could be reduced by partialshading of the bubble column photobioreactor(Kojima and Zhang, 1999). Biomass produc-tion could be doubled under continuous illumi-nation (24 h) when compared with cultures thatexperienced a diurnal light cycle (12 h light/12 hdark); however, compared with diurnal illumi-nation cycle, continuous light resulted in a four-fold increase in hydrocarbon production (Lupiet al., 1994).

Colony size increased with increased lightintensity initially when the cell concentrationwas low and sufficient light for photosynthe-sis was available. As the cell concentrationincreased, the average light intensity withinthe photobioreactor decreased because ofmutual shading (Mirón et al., 1999; Molina etal., 1999, 2000), leading to decrease in colonysize. The production rates of extracellularpolysaccharides and hydrocarbons decreasedwith decreasing average light intensity. It hasbeen suggested that the equilibrium colonysize is determined by a dynamic balance be-tween the mechanical strength of colonies andthe hydrodynamic stress due to turbulence inthe reactors (Zhang and Kojima, 1998; Chisti,1999).

The pH of the culture medium is generallyadjusted to between pH 7.4 and 7.6 beforeinoculation. A regular increase in pH is ob-served during active growth followed by a slightdecline later (Casadevall et al., 1985). The in-crease in pH is partly due to the consumption

Page 24: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

268

of dissolved CO2 for photosynthesis. Similarchanges in pH are commonly observed in CO2-enriched cultures during exponential growth.The optimum temperature for growth is 25°C(Lupi et al., 1991). Sodium fluoride is found tostimulate growth when added at a concentra-tion of 0.1 g·m–3 (Wang and Xie, 1996).

The biomass yield and the cellular compo-sition of B. braunii race A are affected by thesalinity (NaCl concentration) of the culturemedium. A slight increase in the lipid contentwas observed as a result of increasing salinityuntil the salinity equaled that of seawater. Lowerthan normal content of protein, carbohydrates,and pigments were found in cells grown at 0.5 MNaCl (Ben-Amotz et al., 1985). Althoughthe total lipid content of cells remained con-stant, the composition of lipids changed withNaCl concentration. In the lipid fraction theamount of hydrocarbons, acylglycerol, andphosphorus remained constant, but the hexosecontent (especially galactose) decreased. Thiswas attributed to the consumption of UDP-galactose in the biosynthesis of osmoregula-tory compounds. There was also a change inthe fatty acid distribution in polar lipids, andthe proportion of polyunsaturated fatty acidswas higher than the saturated fatty acids whenthe salinity increased. The most important ef-fect of saline stress was a proportional increaseof α-laminaribose (o-β-D-glucopyranosyl-[1-3]-α-D-glucopyranose) content, which act as anosmoprotectant. The concentration ofα-laminaribose tends to be lower in theGottingen strain than in the Austin strain of thealga, and this explains the higher salt toleranceof Austin strain. Although organic osmoregu-latory compounds (e.g., glycerol, glycine) areknown to occur in algae, the osmoprotectoryactivity of α-laminaribose was first reported inB. braunii (Vanquez and Bertha, 1991).

Potentially, domestic sewage that has beenpretreated by activated sludge treatment can beused as a medium for hydrocarbon productionby B. braunii. Using sewage can reduce thecost of producing the hydrocarbons. Secondarystage-treated sewage (STS) has been character-

ized, and found to contain a large amount ofnitrogen (as nitrate) and phosphorus (as PO4

3–).When B. braunii was cultured on STS aeratedwith 1% CO2 at 25°C and without controllingthe pH, the nitrate content could be reducedfrom 7.67 g·m–3 to a level below detection(<0.01 g·m–3). The alga was able to consumePO4

3– present at quite low levels (0.02 g·m–3).NO2

– appeared to be consumed after NO3–, but

NH4+ was not utilized. The growth rate was

0.35 kg·m–3 per week and the hydrocarboncontent was 53% compared with 58% in modi-fied Chu-13 medium under the same condi-tions (Sawayama et al., 1992).

The use of B. braunii for tertiary treatmentof municipal sewage and the conversion of thebiomass to energy can reduce global warmingand eutrophication. Botryococcus cannot growon industrial wastewater containing a high con-centration of inorganic ions. Attempts have beenmade to develop continuous biomass produc-tion processes using secondary-stage treatedwastewater as the culture medium. A 2-l con-tinuous flow bioreactor operated at a dilutionrate of 0.57 d–1 attained a biomass productivityof 196 g·m–3 per week. The bioreactor wasaerated with 1% CO2-enriched air (500 ml/min)and illuminated continuously at 68 µE·m–2s–1.Under these conditions, nitrates and phosphatesdecreased from 5.5 g·m–3 and 0.08 g·m–3 to 4.0g·m–3 and 0.03 g·m–3, respectively (Sawayamaet al., 1994). Potentially, the productivity ofcontinuous algal culture can be improved byoptimizing the dilution rate. The optimal dilu-tion rate is expected to depend on the strengthof wastewater and the intensity of illumina-tion. Productivity can be enhanced further byculturing the cells at an optimal light–darkcycling frequency (Molina et al., 1999, 2000,2001).

B. braunii grew well and was the dominantspecies in mixed cultures of other bacteria and algae(specially Oscillatoria) when cultivated on swinewastewater in an outdoor photobioreactor. At 25°C,the removal rates of chemical oxygen demand(COD), the total dissolved carbon (TDC), total ni-trogen and phosphorus were 0.83, 0.61, 0.69, and

Page 25: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

269

a small scale (Casadevall et al., 1985; Bailliez etal., 1988). Maximum fixation of CO2 and hydro-carbon production by the alga were reported in abubble column photobioreactor. The vessel (7 cmdiameter) was constructed of glass, and it had aconical bottom. The culture was aerated with 1%CO2-enriched sterile air at a rate of 0.5 vvm.Illumination was provided using halogen lampsinstalled on one side of the reactor. The lightintensity was 10 klx (approximately 140 µE·m–2s–

1). The reactor could be partly shaded by an alu-minum foil wrapped on the upper part. Thisachieved a certain light–dark cycling frequency(Molina et al., 1999, 2000, 2001; Mirón et al.,1999), to attempt to relieve photoinhibition(Casadevall et al., 1985). A biomass concentra-tion of 7 kg·m–3 was attained, and the hydrocar-bons constituted 50% of dry cell mass. Bubblecolumns have been claimed to subject the cells toa lesser hydrodynamic stress than similarly con-figured airlift photobioreactors (Kojima andZhang, 1999), but this is debatable.

Compared with closed aseptic culture ves-sels, open pond reactors can provide a moder-ate surface-to-volume ratio at a much lowercost per unit volume (Weissman et al., 1988).However, the culture conditions in opens sys-tems are less well controlled than in closedphotobioreactors and, consequently, the biom-ass productivity is low compared with closedreactors. Also, open culture systems inevitablycontain mixed populations and not just thedesired alga. Notwithstanding these problems,open culture systems such as the “raceway”ponds may be the most realistic for producinglarge amounts of B. braunii for extracting hy-drocarbons. This would be so especially if aninexpensive source of CO2 could be found tocircumvent the relatively low CO2 utilizationefficiency of open channels and ponds.

VIII. USE OF IMMOBILIZED ALGALCELLS

Use of immobilized cell culture offers im-portant advantages over free suspension cul-ture especially when the cells are slow grow-

0.16 g·m–3 per day, respectively. The lipid content ofthe alga grown in swine wastewater and the modi-fied Chu-13 medium were 32.8 ± 3.2% and 33.2 ±2.6%, respectively (Lee et al., 1999).

B. Photobioreactors Used forCultivation

Many kinds of photobioreactors are avail-able for culturing microalgae (Molina et al.,1999; Mirón et al., 1999, 2000), and most ofthese have been evaluated for B. braunii cul-ture. Batch and continuous schemes of opera-tion have been tested in open and closed (axenic)bioreactors. Because of economics, only con-tinuous culture is realistically feasible for thelarge-scale production of microalgal biomass.A commercially viable continuous culture pro-cess needs to attain a high density of biomass,or the biomass productivity will be unaccept-ably low. Because light penetration in densecultures of microalgae is extremely limited,only thin channel flat plate photobioreactorsand narrow bore tubular bioreactors are likelyto be satisfactory for commercial production.The channel depth and tube diameter in suchreactors would be generally ≤10 cm. Althoughthin channel flat plate types of photobioreactorsare known to be highly efficient, they are noteasily scaled up to sizes that will be necessaryfor producing significant quantities of hydro-carbons from B. braunii. This narrows thechoice to tubular photobioreactors. Generally,tubular photobioreactors have provided highbiomass productivities in closed cultures ofmicroalgae (Molina et al., 1999; AciénFernández et al., 2001). Tubular bioreactorsrequire a large land area for a given volume ofreactor, and this is a significant disadvantage.

Bubble columns and airlift reactors that aremore compact than tubular devices can offerimportant advantages for large scale culture if acertain loss of productivity is accepted (Mirón etal., 1999, 2000). Continuous culture in airliftbioreactors (Chisti, 1989; Acién Fernández et al.,2001; Mirón et al., 1999, 2000) has been usedespecially frequently for producing B. braunii at

Page 26: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

270

ing. Extracellular products can be recoveredcontinuously from immobilized cell systems.Immobilization allows reuse of the biomassand eases separation of biomass from the sus-pending fluid. Also, immobilization can pro-tect cells against hydrodynamic shear forces,and this can be important because microalgaecan be shear-sensitive (Chisti, 1989, 1999). Inview of these factors, many immobilizationschemes have been examined for use withB. braunii cultures.

Immobilization in calcium alginate beadsresults in cells with enhanced chlorophyll con-tent and photosynthetic activity relative to freecontrols. This behavior is seen at all stages ofa batch culture under continuous illumination.Immobilization reduces the irradiance seen bythe cells, and they respond by enhancing thechlorophyll content in attempts to capture moreof the available light. Also, the gel-entrappedcells are protected from photoinhibition, a con-dition in which intense irradiance actuallycauses a loss in photosynthetic performance(Molina et al., 1999; Mirón et al., 1999).

Alginate gel-immobilized cells have lowergrowth rates and lower biomass productionrelative to free controls. This is possibly be-cause immobilization can reduce access ofnutrients to the cells and the immobilizationmatrix has a colony-confining effect. Immobi-lized cells retain the ability to produce hydro-carbons, whose structure and relative abun-dance are not affected by immobilization(Bailliez et al., 1986). In some cases, a notableincrease in the hydrocarbon production (+20%compared with free cell system) has been re-corded for immobilized cells, suggesting im-mobilization-related diversion of metabolicpathway toward hydrocarbon synthesis (Bailliezet al., 1985).

Immobilization by occlusion in polyure-thane foams (PUF) has been used for B. brauniicells. PUF support matrices are versatile be-cause of their broad range of porosities andmechanical properties. The immobilization ofB. braunii cells in PUF via entrapment has ledto reduced photosynthetic activity and hydro-

carbon production in some cases. Nutrient anddiffusion limitations are apparently not the causeof reduced performance (Bailliez et al., 1985),but light limitation is a plausible explanation.A study of PUF-immobilized Anabaenavariabilis associated the observed poor perfor-mance with the toxic nature of the matrix. PUFmatrix can release water-soluble toxic com-pounds (Gudin and Thomas, 1981). Also, in-creased local temperature and pressure in thematrix may explain some of the poor results.Some foam-matrix immobilized cultures canexperience a substantial leakage (e.g., ~30%)of cells (Bailliez et al., 1988). One study ob-served that the hydrocarbon production of foam-adsorbed cultures was comparable to that offreely suspended cells when cultivated in aairlift photobioreactor for 3 weeks (Bailliez etal., 1988). The average size of the foam-adsorbed colonies remained quite small andallowed greater recovery of hydrocarbons fromthe culture by solvent extraction (Bailliez et al.,1988).

In one case, cotton gauze-immobilizedB. braunii cells showed higher levels of hydro-carbon production, biomass growth, and pho-tosynthetic activity when compared with cellsimmobilized in PUF (Yang and Wang, 1989).Despite advantages, the practicability of im-mobilized algal culture remains questionableunless the cells can be cultured in long-dura-tion continuous culture and the hydrocarbonscan be extracted continuously.

IX. DOWNSTREAM PROCESSING

A. Hydrocarbon Recovery

Product recovery from fermentation andcell culture systems typically requires a seriesof unit operations that often control the eco-nomic viability of a production process (Chisti,1998). The selection and design of a recoveryscheme is related with the nature of the sourceorganism and the properties of the product.Producing the biomass at a low cost can have

Page 27: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

271

a profound impact on the overall cost of prod-uct recovery (Belarbi et al., 2000), especially ifthe product does not need to be purified exten-sively.

Once the B. braunii biomass has been re-covered from the broth by filtration or centrifu-gation, the hydrocarbons can be recovered ei-ther by pressing out or by the solvent extractionof the biomass. Pressing out the oils has beenattempted on a laboratory scale, but no infor-mation is available on the extent of recovery.Because B. braunii has a thick cell wall and thewet biomass paste contains 10 times more wa-ter than hydrocarbons, recovery by pressing isnot efficient. Also, this recovery method can-not be applied to continuous culture of immo-bilized cells.

Solvent extraction of biomass can removethe hydrocarbons without damaging the cells,if the solvent used is nontoxic. In addition,the selected solvent should have the follow-ing properties: the solvent should be immis-cible with water; it should preferentially solu-bilize the product of interest; it should havea low boiling point for ease of removal; itshould have a density that is significantlydifferent than water; it should be readily avail-able and inexpensive; and it should be reus-able. In view of these considerations, hexaneappears to be the solvent of choice for ex-tracting algal hydrocarbons (Frenz et al.,1989a, b). For efficient extraction, the cellsmust first be concentrated to a paste becausethe presence of too much water around thecells reduces contact of the nonpolar solventwith the outer cell wall where the hydrocar-bons are stored.

Under suitable conditions, 70% of hydro-carbons can be released by 30 min of contactwith hexane. Growth and hydrocarbon produc-tion are not affected by repeated extractionwith hexane. In fact, a higher content of hydro-carbons has been observed in hexane-treatedbiomass relative to controls. Recovery yieldsare influenced by physiological status of cul-ture. In one case, cells in the early exponentialgrowth phase permitted greater recovery of

hydrocarbons when cultured in a airliftphotobioreactor compared with culture in stan-dard stirred bioreactor (Frenz et al., 1989a).This effect was explained by the smaller aver-age size of colonies in the stirred vessel (Frenzet al., 1989a). The smaller average colony sizein the stirred tank was probably caused by ahigher turbulence in the tank than in the airliftreactor (Chisti, 1999).

Scale up of extraction can be difficult, be-cause the alga tends to aggregate and formclumps during extraction. This happens becauseof differences in polarities of the solvent andthe wet cells. Clumping shields a large fractionof the biomass from exposure to solvent. Agi-tation at high speed can overcome this prob-lem, but intense agitation damages the cells(Chisti, 1999). Also, intense agitation can bequite expensive for large-scale commercialoperations.

Hydrocarbons can be recovered continu-ally from immobilized cells. Recovery yieldsare markedly increased relative to freely sus-pended controls when the cells are immobi-lized by adsorption in polyurethane foam (PUF).Because the colonies are at the surface of theimmobilization matrix, they are easily accessedby the solvent. Optimal contact time was foundto be 30 min when extracting with hexane (Frenzet al., 1989a, b).

Supercritical fluid (SCF) extraction is an-other technology that has been applied toB. braunii (Mendes et al., 1995). Because ofrelatively low values of viscosity, density,and surface tension, supercritical fluids suchas CO2 allow rapid extraction of solids.Supercritical CO2 is nontoxic, inexpensive,easily removed from the extract, and reus-able. A relatively low value of critical tem-perature allows use of CO2 for extractingthermolabile compounds. In the extraction ofalgal hydrocarbons with CO2 at 30°C, thesolubility of hydrocarbons was observed toincrease with increasing pressure (Mendes etal., 1995). The highest rate of extraction wasobtained at a pressure of 30 MPa (Mendes etal., 1995).

Page 28: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

272

B. Converting Algal Hydrocarbonsand Oil-Rich Biomass to Fuels

Hydrocarbons obtained in the hexane ex-tract of B. braunii can be burnt directly; how-ever, for improved performance in internalcombustion engines, the oil must be modifiedby processes such as pyrolysis and catalyticcracking.

Wet and dry algal biomass can be sub-jected to direct thermochemical liquefaction toproduce combustible oil. Thermochemical liq-uefaction for 1 h (300°C, 10 MPa pressure) inthe presence of 5% Na2CO3 as a catalyst pro-vides a good yield of oil (Sawayama et al.,1999). This oil can be fractionated by silica gelcolumn chromatography into three fractions,as follows: (1) a low-molecular weight (197 to281 Da) fraction formed by the degradation ofbotryococcenes (C17–C22 hydrocarbons), con-stituting about 5% of total; (2) Botryococceneswith molecular weights of 438 to 572 Da, con-stituting 27.2% of the oil; (3) Polar substances(867 to 2209 Da molar mass) produced fromorganic material other than hydrocarbons andconstituting 22.2% of the oil (Inoue, 1994).Thermochemical liquefaction of biomass elimi-nates the need for initial solvent extraction forrecovery of hydrocarbons. However, the over-all yield of oil from liquefied biomass is low(52.9%) compared with yield obtained by hex-ane extraction (Dote et al., 1992). Of course,the thermochemical processing of biomass isnot applicable to continuous culture operationswhere all biomass is retained in the culturevessel and hydrocarbon extraction occurs con-tinually in situ (Frenz et al., 1989a, b).

Crude hydrocarbons of B. braunii can beconverted to gasoline (60 to 70%), light cycleoil (10 to 15%), heavy cycle oil (2 to 8%), andcoke (5 to 10%) by subjecting the crude oil tocatalytic cracking (Kitazato et al., 1989). Avariety of cracking catalysts can be used, in-cluding Co-Mo catalyst (Held et al., 1985) andzeolites (Kitazato et al., 1989). Yield of gaso-line and other fractions depend on the catalystand the reaction temperature. The optimal con-

version temperature with zeolite catalyst is497°C. The yields of gasoline obtained by cata-lytic cracking of algal hydrocarbons are com-parable to yields obtained from petroleum. Also,the gasoline produced has a sufficiently highoctane number for direct use in automobiles.High octane numbers result because of the for-mation of alkylbenzenes, mainly xylenes andtrimethyl-benzenes. These aromatic hydrocar-bons are derived from botryococcenes (mainlythe C34H58 compound) directly through con-secutive cracking or cyclo-aromatization(Kitazato et al., 1989). Figure 6 provides amechanistic representation of catalytic crack-ing of botryococcene, C34H58. A highly specificoxidation of the vinyl double bond inbotryococcene (C34H58) has been used to con-vert this compound to botryococcone, C34H58O,a methyl ketone (Chisti, 1980).

X. CLONING OF THE HYDROCARBONGENES

Only a few reports exist on the cloning ofB. braunii genes into other organisms. Recentlya 366-bp cDNA fragment was obtained from theB race using reverse transcriptase/polymerasechain reaction (PCR) and degenerate primersbased on conserved amino acid sequences foundin all squalene synthase enzymes. Using thatputative squalene synthase fragment as a probe,a 2632-bp cDNA clone was isolated from acDNA library. The cDNA contained an openreading frame (ORF) coding for a protein with461 amino acids and a molecular mass of 52.5kDa. A comparison of Botryococcus squalenesynthase (BSS) gene with that from other organ-isms showed a 52% homology with Nicotianatabacum, 51% with Arabidopsis thaliana, 48%with Zea mays, 40% with rat, 39% with Saccha-romyces cerevisiae, and 26% with Zymomonasmobilis (Okada et al., 2000).

The entire ORF region of BSS was ampli-fied by PCR using a forward primer havingNdeI restriction site and a start codon and areverse primer having BclI site and stop codon

Page 29: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

273

to generate a 1403-bp fragment. A carboxyterminal truncated BSS of 1325 bp was alsogenerated by PCR using the same forwardprimer and different reverse primer. The ex-pression of full-length and carboxy terminaltruncated BSS in E. coli BL21 in presence ofisopropyl thio-β-D-galactoside (IPTG) resultedin significant levels of squalene synthase en-zyme activity, but no botryococcene synthaseactivity.

Squalene synthase is an endoplasmic reticu-lum-associated enzyme with the carboxy ter-minus tethering the enzyme to the ER mem-brane (Robinson et al., 1993). The associationwith ER hampers the conventional purificationof the enzyme. RNA blot hybridization analy-sis of algal cultures indicated preferential ex-pression of BSS gene during rapid growth.Southern blot analysis indicated a single copyof SS gene in the algal genome (Okada et al.,2000). All these results suggest that either co-ordinated expression of separate synthase genesfor squalene and botryococcenes exists or thatthere are physiological conditions controlling

the squalene synthase vs. botryococcene syn-thase activity of a single polypeptide species.

XI. CONCLUDING REMARKS

In view of the material reviewed, B. brauniiis a potentially good renewable source of use-ful hydrocarbons, polysaccharides, and otherspecialty chemicals. Catalytically cracked al-gal hydrocarbons have sufficiently high octaneratings for use as motor fuel. The use of algalhydrocarbons can greatly reduce the environ-mental impact associated with using coal andpetroleum. Also, B. braunii can be used forremoving low levels of contaminating nitratesand phosphates from wastewater.

At present, the production of photosyntheticfuel oils is not competitive with petroleum-derived fuels. One reason for this is a relativelyslow growth rate of B. braunii. The hydrocar-bon synthesis genes of B. braunii have beencloned into other microorganisms. This canpotentially increase the production rates and

FIGURE 6. Production of aromatic hydrocarbons by catalyticcracking of a typical botryococcene, C34H58.

Page 30: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

274

reduce problems associated with a high viscos-ity of B. braunii broths. Substantial develop-mental effort is needed to consistently cultureB. braunii in large-scale systems.

REFERENCES

Acién Fernández, F. G., Fernández Sevilla, J. M.,Sánchez Pérez, J. A., Molina Grima, E., andChisti, Y. 2001. Airlift-driven external-loop tu-bular photobioreactors for outdoor production ofmicroalgae: assessment of design and perfor-mance. Chem. Eng. Sci. 56: 2721.

Allard, B. and Casadevall, E. 1990. Carbohydrate com-position and characterisation of sugars from thegreen microalga Botryococcus braunii. Phytochem.29: 1875.

Anderson, D. B. and Eakin, D. E. 1985. A process for theproduction of polysaccharides from microalgae.Biotechnol. Bioeng. Symp. 15: 533.

Bailliez, C., Largeau, C., and Casadevall, E. 1985. Growthand hydrocarbon production of Botryococcusbraunii immobilized in calcium alginate gel. Appl.Microbiol. Biotechnol. 23: 99.

Bailliez, C., Largeau, C., Berkaloff, C., and Casadevall,E. 1986. Immobilization of Botryococcus brauniiin alginate, influence on chlorophyll content,photosynthetic activity and degeneration duringbatch culture. Appl. Microbiol. Biotechnol. 23:361.

Bailliez, C., Largeau, C., Casadevall, E., Yang, L. W.,and Berkaloff, C. 1988. Photosynthesis, growthand hydrocarbon production of Botryococcusbraunii immobilized by entrapment and adsorp-tion in polyurethane foams. Appl. Microbiol.Biotechnol. 29: 141.

Behrens, A., Schaeffer, P., Bernasconi, S., and Alrecht,P. 2000. 7,11-cyclobotryococca 5,12,26-triene,a novel botryococcene related hydrocarbon oc-curring in environment. Organic Lett. 2: 1271.

Belarbi, E. H., Molina, E., and Chisti, Y. 2000. Aprocess for high yield and scaleable recovery ofhigh purity eicosapentaenoic acid esters frommicroalgae and fish oil. Enzyme Microb. Technol.26: 516.

Ben-Amotz, A., Torbene, T. G., and Thomas, W. H.1985. Chemical profile of selected species of

microalgae with emphasis on lipids. J. Phycol.21: 72.

Bertheas, O., Metzger, P., and Largeau, C. 1999. Ahigh molecular weight complex lipid, aliphaticpolyaldehyde tetraterpene diol polyacetal fromrace L of Botryococcus braunii. Phytochemistry50: 85.

Blackburn, K. B. 1936. Botryococcus and algal coal.Trans. R. Soc. Edin. 58: 841.

Brenckman, F., Largeau, C., Casadevall, E., andBerkaloff, C. 1989. Effect of nitrogen nutritionon growth and hydrocarbon production of theunicellular microalga Botryococcus braunii.Comm. Eur. Communities, Energy Biomass 717.

Brown, A. G. and Knights, B. A. 1969. Hydrocarboncontent and it’s relationship to physiological statein the green alga Botryococcus braunii. Phy-tochemistry 8: 543.

Cane, R. F. 1977. Coorongite, balkashite and relatedsubstances—an annotated bibliography. Trans.R. Soc. S. Aust. 101: 153.

Casadevall, E., Largeau, C., Metzger, P., Chirac, C.,Berkaloff, C., and Coute, A. 1983. Hydrocarbonproduction by unicellular microalga Botryococcusbraunii. Biosciences 2: 129.

Casadevall, E., Dif, D., Largeau, C., Gudin, C.,Chamount, D., and Desanti, O. 1985. Studies onbatch and continuous culture of Botryococcusbraunii: hydrocarbon production in relation tophysiological state, cell ultrastructure and phos-phate nutrition. Biotechnol. Bioeng. 27: 286.

Chirac, C., Casadevall, E., Largeau, C., and Metzger,P. 1985. Bacterial influence upon growth andhydrocarbon production of the alga Botryococcusbraunii. J. Phycol. 21: 380.

Chisti, Y. 1980. An unusual hydrocarbon. J. RamsaySociety 27–28: 24–26.

Chisti, Y. 1989. Airlift Bioreactors, Elsevier, London.

Chisti, Y. 1998. Strategies in downstream processing.In Bioseparation and Bioprocessing: A Hand-book, vol. 2, Subramanian, G., Ed., Wiley-VCH,New York, 3.

Chisti, Y. 1999. Shear sensitivity. In: Encyclopediaof Bioprocess Technology: Fermentation,Biocatalysis, and Bioseparation, vol. 5,Flickinger, M. C. and Drew, S. W., Eds., Wiley,New York, 2379.

Page 31: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

275

David, M., Metzger, P., and Casadevall, E. 1988. Twocyclobotryococcenes from the B race of greenalga Botryococcus braunii. Phytochemistry 27:2863.

Dennis, M. W. and Kolattukudy, P. E. 1991. Alkanebiosynthesis by decarbonylation of aldehydecatalyzed by a microsomal preparation fromBotryococcus braunii. Arch. Biochem. Biophys.287: 268.

Derenne, S., Largeau, C., Casadevall, E., and Connan,J. 1988. Mechanism of formation and chemicalstructure of coorongite. Structure and origin ofthe labile fraction. Fate of botryococcenes dur-ing early diagenesis. Org. Geochem. 13: 965.

Dernne, S., Largeau, C., Casadevall, E., and Berkaloff,C. 1989. Comparative study of rat and tobaccosqualene. Phytochemistry 38: 1137.

Dote, Y., Sawayama, S., and Yokoyama, S. 1992.Liquefaction of hydrocarbon-rich microalga. Am.Chem. Soc. Div. Fuel Chem. 37: 1836.

Fallon, R. D. and Brock, T. D. 1979. Decomposition ofblue green algal (cyanobacterial) blooms in lakeMendota, Wisconsin. Appl. Environ. Microbiol.37: 820.

Fernandes, H. L., Tomme, M. M., Lupi, F. M., Fialho,A. M., Sa Correia, I., and Novais, J. M. 1989.Biosynthesis of high concentration of anexopolysaccharide during cultivation of themicroalga Botryococcus braunii. Biotechnol. Lett.11: 433.

Frenz, J., Largeau, C., and Casadevall, E. 1989a. Hy-drocarbon recovery by extraction with abiocompatible solvent from free and immobi-lized cultures of Botryococcus braunii. EnzymeMicrob. Technol. 11: 717.

Frenz, Z., Largeau, C., Csadevall, C., Kollerup, F., andDaugulis, A. J. 1989b. Hydrocarbon recovery andbiocompatibility of solvents for extraction fromcultures of Botryococcus braunii. Biotechnol.Bioeng. 34: 755.

Gelpi, E., Schneider, H., Mann, J., and Oro, J. 1970.Hydrocarbons of geochemical significance inmicroscopic algae. Phytochemistry 9: 603.

Goodway, G. W., Fawcett, P., Green, D., and Shaw, G.1973. Sporopollenin formation in the ascospore wallof Neurospora crassa. Arch. Microbiol. 101: 145.

Grung, M., Metzger, P., and Liaaen-Jensen, S. 1989.Algal carotenoids, primary and secondary caro-

tenoids in two races of green alga Botryococcusbraunii. Biochem. Sys. Eco. 17: 263.

Grung, M., Metzger, P., and Liaaen-Jensen, S. 1994.Secondary carotenoids in Botryococcus brauniirace L, new strain. Biochem. Sys. Eco. 22: 25.

Gu, P., Ishii, Y., Spencer, T. A., and Shechter, I. 1998.Function-structure studies and identification ofthree enzyme domain involved in the catalyticactivity in rat hepatic squalene synthase. J. Biol.Chem. 273: 12515

Gudin, C. and Thomas, D. 1981. Production de polysac-charides sulfates par un biophotoreacteur a cel-lules immobilisees de Porphyridium curentum.C. R. Sci. Paris 293: 35.

Gudin, C., Bernard, A., Chaumont, D. T., Thepnier,C., and Hardy, T. 1984. Direct conversion ofsolar energy into organic chemicals. WorldBiotech. Rep. 1: 541.

Guzman-Murillo, M. A. and Ascencio, F. 2000. Antiadhesive activity of sulfated polysaccharides ofmicroalgae on attachment of red sore disease-associated bacteria and Helicobacter pylori totissue culture cells. Lett. Appl. Microbiol. 30:473.

Haines, K. C. and Guillard, R. R. L. 1974. Growth ofvitamin B12 requiring marine diatoms in mixedlaboratory cultures with vitamin B12 producingmarine bacteria. J. Phycol. 10: 245.

Held, W., Peters, M., Buhs, C., Oelert, H. H.,Reifenstahl, G., and Wagner, F. 1985. Produc-tion of hydrocarbon from biomass. Comm. Eur.Communities, Energy Biomass 744.

Huang, Z., Poulter, C. D., Wolf, F. R., Somers, T. C.,and White, J. D. 1988. Braunicene, a novel cy-clic C32 isoprenoid from Botryococcus braunii.J. Am. Chem. Soc. 110: 3959.

Huang, J. and Poulter, C. D. 1989. Tetramethylsqualene, a triterpene from Botryococcus brauniivar showa. Phytochem. 28: 1467.

Inone, H., Korenaga, T., Sagami, H., Koyama, T.,Sugiyama, H., and Ogura, K. 1993. Formation offarnesal and 3-hydroxy-2,3-dihydrofarnesal fromfarnesol by protoplasts of Botryococcus braunii.Biochem. Biophys. Res. Commun. 196: 1401.

Inone, H., Korenaga, T., Sagami, H., and Ogura, K.1994a. Phosphorylation of farnesol by a cell freesystem of Botryococcus braunii. Biochem.Biophys. Res. Commun. 200: 1036.

Page 32: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

276

Inoue, S., Dote, Y., Sawayama, S., Minowa, T., Ogi,T., and Yokoyama, S. 1994b. Analysis of oilderived from liquefaction of Botryococcusbraunii. Biomass Bioenergy 6: 269.

Jarstfer, M. B., Blagg, B. S. J., Rogers, D. H., andPoulter, C. D. 1996. Biosynthesis of squalene.Evidence for a tertiary cyclopropylcarbinyl cat-ionic intermediate in the rearrangement ofpresqualene diphosphateto squalene. J. Am.Chem. Soc. 118: 13089.

Jones, J. G. 1972. Studies on freshwater bacteria: as-sociation with algae and alkaline phosphataseactivity. J. Ecol. 60: 59.

Kitazato, H., Asaoka, S., and Iwamoto, H. 1989. Cata-lytic cracking of hydrocarbons from microalgae.Sekiyu Gakkaishi 32: 28.

Kojima, E. and Zhang, K. 1999. Growth and hydrocar-bon production by microalga Botryococcusbraunii in bubble column photobioreactor. J.Bioscience Bioeng. 87: 811.

Koren, K. M. and Rayburn, W. R. 1984. Influence ofgrowth status and nutrients on exocellularpolysaccharides synthesis by the soil algaChlamydomonas mexicana. J. Phycol. 20: 253.

Lareillard, J., Largeau, C., and Casadevall, E. 1988.Oleic acid in the biosynthesis of resistant poly-mer of Botryococcus braunii. Phytochemistry27: 2095.

Largeau, C., Casadevall, E., Berkaloff, C., andDhamliencourt, P. 1980. Sites of accumulationand composition of hydrocarbons in Botryococcusbraunii. Phytochemistry 19: 1043.

Lee, S. J., Kim, S. B., Kim, H. S., Kwon, G. S., Yoon,B. D., and Oh, H. M. 1999. Mass cultivation ofBotryococcus braunii for advanced treatment ofswine waste water and lipid production inphotobioreactor. Sanop Misaengmul Hakhoechi27: 166.

Lichitfouse, E., Derenne, S., Mariotti, A., and Largeau,C. 1994. Possible algal origin of long chain oddn-alkanes in immature sediments as revealed bydistribution and carbon isotope ratios. Org.Geochem. 22: 1023.

Lupi, F. M., Fernandes, H. M. L., Sa Correia, I., andNovais, J. M. 1991. Temperature profiles of cel-lular growth exopolysaccharide synthesis byBotryococcus braunii. J. Phycol. 3: 35.

Lupi, F. M., Fernandes, H. M. L., Tomme, M. M., SaCorreia, I., and Novais, J. M. 1994. Influence ofnitrogen source and photoperiod on exopolysaccharidesynthesis by the microalga Botryococcus braunii.Enzyme Microb. Technol. 16: 546.

Mcirdy, D. M., Cox, R. E., Vlkman, J. K., and Howel,V. J. 1986. Botryococcane in a new class ofAustralian non marine crude oils. Nature 320:57.

Mendes, R. L., Coelho, J. P., and Fernandes, H. L.1995. Application of supercritical CO2 extrac-tion to microalgae and plants. J. Chem. Technol.Biotechnol. 62: 53.

Metzger, P., Berkaloff, C., Casadevall, E., and Coute,A. 1985a. Alkadiene and botryococcene produc-ing races of wild strains of Botryococcus braunii.Phytochem. 24: 2305.

Metzger, P., Casadevall, E., Pouet, M. J., and Pouet, Y.1985b. Structure of some botryococcenes:branched hydrocarbons from the B race of thegreen alga Botryococcus braunii. Phytochem.24: 2995.

Metzger, P. and Casadevall, E. 1987. Lycopadiene, atetraterpenoid hydrocarbon from new strains ofthe green alga Botryococcus braunii. Tetrahe-dron Lett. 24: 2305.

Metzger, P., David, M., and Casadevall, E. 1987. Bio-synthesis of triterpenoid hydrocarbons in the Brace of green alga Botryococcus braunii. Sites ofproduction and nature of methylating agent.Phytochem. 26: 129.

Metzger, P., Casadevall, E., and Coute, A. 1988.Botryococcene distribution in strains of greenalga Botryococcus braunii. Phytochemistry 27:1383.

Metzger, P. and Casadevall, E. 1989. Aldehydes, verylong chain alkenyl phenols, epoxides and otherlipids from an alkadiene producing strain ofBotryococcus braunii. Phytochemistry 28: 2097.

Metzger, P., Allard, B., Casadevall, E., Berkaloff, C.,and Coute, A. 1990. Structure and chemistry ofa new chemical race of Botryococcus braunii(chlorophyceae) that produces lycopadiene, atetraterpenoid hydrocarbon. J. Phycol. 26: 258.

Metzger, P. and Casadevall, E. 1991. Botryococcoidethers, ether lipids from Botryococcus braunii.Phytochemistry 30: 1439.

Page 33: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

277

Metzger, P., Villarreal-Rosales, E., and Casadevall, E.1991. Methyl branched fatty aldehydes and fattyacids in Botryococcus braunii. Phytochemistry30: 185.

Metzger, P. and Casadevall, E. 1992. Ether lipids fromBotryococcus braunii and their biosynthesis.Phytochemistry 31: 2341.

Metzger, P. and Pouet, Y. 1995. n-Alkyl pyrogalloldimethyl ethers, aliphatic diol monoesters andsome minor ether lipids from Botryococcusbraunii (A race). Phytochemistry 40: 543.

Metzger, P. and Aumelas, A. 1997. Lycopanerol-A,di-tetraterpenoid tetraether derivatives from greenmicroalga Botryococcus braunii L race. Tetra-hedron Lett. 38: 2977.

Mirón, A. S., Gómez, A. C., Camacho, F. G., Grima, E.M., and Chisti, Y. 1999. Comparative evaluationof compact photobioreactors for large-scale mo-noculture of microalgae. J. Biotechnol. 70: 249.

Mirón, A. S., Camacho, F. G., Gómez, A. C., Grima,E. M., and Chisti, Y. 2000. Bubble column andairlift photobioreactors for algal culture. AIChEJ. 46: 1872.

Moldwan, J. M. and Seifert, W. K. 1980. First discov-ery of botryococcane in petroleum. J. C. S. Chem.Commun. 1: 912.

Molina, E., Fernández, F. G. A., Camacho, F. G., andChisti, Y. 1999. Photobioreactors: light regime,mass transfer, and scaleup. J. Biotechnol. 70:231.

Molina, E., Acién Fernández, F. G., García Camacho,F., Camacho Rubio, F., and Chisti, Y. 2000.Scale-up of tubular photobioreactors. J. Appl.Phycol. 12: 355.

Molina, E., Fernández, J., Acién, F. G., and Chisti, Y.,2001. Tubular photobioreactor design for algalcultures. J. Biotechnol. 92: 113.

Murakami, M., Nakano, H., Yamaguchi, K., Konosu, S.,Nakayama, O., Matsumoto, Y., and Iwamoto, H.1988. Meijicoccene, a new cyclic hydrocarbon fromBotryococcus braunii. Phytochemistry 27: 455.

Murakami, N. 2000. Plant growth regulator composi-tion and their manufacture. Jpn. Kokai TokkyoKoho 264: 809.

Murry, J. and Thomson, A. 1977. Hydrocarbon pro-duction in Anacystis montana and Botryococcusbraunii. Phytochemistry 16: 465.

Oh, H. M., Kim, S., Park, E. R., Lee, S. T., Kwon, J.S., and Yoon, B. D. 1997. Effects of light inten-sity and nutrients on the growth of Botryococcussp. Misaengmul Hakhoechi 25: 339.

Ohmori, M., Wolf, F. R., and Bassham, J. A. 1984.Botryococcus braunii carbon/nitrogen metabo-lism as affected by ammonia addition. Arch.Microbiol. 140: 101.

Okada, S., Mastuda, M., and Yamaguchi, K. 1996.Botryoxanthin-A a new member of the newclass of carotenoids from green microalgaBotryococcus braunii, Berkeley. TetrahedronLett. 37: 1065.

Okada, S., Tonegawa, I., Mastuda, H., Murakami, M.,and Yamaguchi, K. 1997. Carotenoids from greenmicroalga Botryococcus braunii. Tetrahedron 53:11307.

Okada, S., Tonegawa, I., Mastuda, H., Murakami, M.,and Yamaguchi, K. 1998. Botryoxanthin-B andalpha botryoxanthin-A from green microalgaBotryococcus braunii. Phytochemistry 47: 1111.

Okada, S., Devarenne, T. P., and Chappel, J. 2000.Molecular characterization of squalene synthasefrom the green alga Botryococcus braunii, raceB. Arch. Biochem. Biophys. 373: 307.

Parker, B. C. and Bold, H. C. 1961. Biotic relation-ships between soil algae and other microorgan-isms. Am. J. Botany 48: 185.

Poulter, C. D. and Hughes, J. M. 1977. Model studiesof the biosynthesis of non-head to tail terpenes.Stereochemistry of ionization for N-methyl-4-[(1S, 1’R, 3’R)-[1-2H] chrysanthemyloxy]-pyridinium iodide. J. Am. Chem. Soc. 99: 3824.

Poulter, C. D., Marsh, L., Argyle, J. C., Dennis, M. S.,Robyn, J. G., and Scott, G. M. 1977. Modelstudies of biosynthesis of non-head to tail terpe-nes. Rearrangements of the chrysanthemyl sys-tem. J. Am. Chem. Soc. 99: 3816.

Rager, M. N. and Metzger, P. 2000. Six noveltetraterpenoid ethers, lycopanerols B–G, andsome other constituents from green microalgaBotryococcus braunii. Phytochemistry 54: 427.

Reim, R. L., Shane, M. S., and Cannon, R. E. 1974. Thecharacterization of a Bacillus capable of blue greenbactericidal activity. Can. J. Microbiol. 20: 981.

Robinson, G. Y., Tsay, Y. H., Kienzle, B. K., Smith-Monroy, C. A., and Bishop, R. W. 1993.

Page 34: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

278

Conservation between human and fungalsqualene synthases: similarities in structure, func-tion and regulation. Mol. Cell Biol. 13: 2706.

Saks, N. M. and Kahn, E. G. 1979. Substrate compe-tition between a salt marsh diatom and a bacte-rial population. J. Phycol. 15: 17.

Sawayama, S., Minowa, T., Dote, Y., and Yokoyama,S. 1992. Growth of the hydrocarbon richmicroalga Botryococcus braunii in secondarilytreated sewage. Appl. Microbiol. Biotechnol. 38:135.

Sawayama, S., Inone, S., and Tokoyama, S. 1994.Continuous culture of hydrocarbon rich microalgaBotryococcus braunii secondarily treated sew-age. Appl. Microbiol. Biotechnol. 41: 729.

Sawayama, S., Inoue, S., and Yokoyama, S. 1995.Phylogenetic position of Botryococcus brauniibased on small subunit of ribosomal RNA se-quence data. J. Phycol. 31: 419.

Sawayama, S., Minowa, T., and Yokoyama, S. 1999.Possibility of renewable energy production andcarbon dioxide mitigation by thermochemicalliquefaction of microalgae. Biomass Bioenergy17: 33.

Swale, E. M. F. 1968. The phytoplankton of Oakmere,Cheshire. Br. Phycol. Bull. 3: 441.

Templier, J., Largeau, C., and Casadevall, E. 1984.Hydrocarbon formation in the green algaBotryococcus braunii and mechanism of non-isoprenoid hydrocarbon biosynthesis. Phytochem.23: 1017.

Templier, J., Largeau, C., and Casadevall, E. 1991a.Biosynthesis of n-alkatrienes in Botryococcusbraunii. Phytochemistry 30: 2209.

Templier, J., Largeau, C., and Casadevall, E. 1991b.Non-specific elongation-decarboxylation in bio-synthesis of cis and trans-alkadienes byBotryococcus braunii. Phytochemistry 30: 175.

Templier, J., Disendorf, C., Largeau, C., and Casadevall,E. 1992a. Metabolism of n-alkadienes in the Arace of Botryococcus braunii. Phytochemistry31: 113.

Templier, J., Largeau, C., and Casadevall, E. 1992b.Chemical inhibition of resistant biopolymers inthe outer walls of A and B races of Botryococcusbraunii. Phytochemistry 31: 4097.

Templier, J., Largeau, C., and Casadevall, E. 1993.Variation in external and internal lipid associ-

ated with inhibition of resistant biopolymer fromthe A race of Botryococcus braunii. Phytochem-istry 33: 1079.

Tonegawa, I., Okada, S., Murakami, M., andYamaguchi, K. 1998. Pigment composition ofthe green microalga Botryococcus braunii,Kawaguchi. Fish Sc. 64: 305.

Vanquez, R. and Bertha, O. 1991. Haloadaption of thegreen alga Botryococcus braunii. Phytochemis-try 30: 2919.

Volova, T. G., Kalacheva, G. S., Zhilo, N. O., andPlotnicov, V. F. 1998. Physiological and bio-chemical properties of the alga Botryococcusbraunii. Russ. J. Plant. Physol. 45: 725.

Wake, L. V. and Hillen, L. W. 1981. Nature and hydro-carbon content of the blooms of the algaBotryococcus braunii occurring in Australian freshwater lakes. Aust. J. Mar. Freshwater Res. 32: 353.

Wang, X. and Kolattukudy, P. E. 1995. Solubilizationand purification of aldehyde generating fatty acyl-CoA reductase from green alga Botryococcusbraunii. FEBS Lett. 370: 15.

Wang, X. and Xie, S. 1996. Effects of several factors ongrowth of Botryococcus braunii. WeishengwuxueTongbao 23: 275.

Weetal, H. H. 1985. Studies on nutritional require-ments of the oil producing alga Botryococcusbraunii. Appl. Biochem. Biotechnol. 11: 377.

Weissman, J. C., Goebel, R. P., and Benemann, J. R.1988. Photobioreactor design: mixing, carbonutilization, and oxygen accumulation. Biotechnol.Bioeng. 31: 336.

Wolf, F. R., Nanomura, A. M., and Bassham, J. A.1985a. Growth and branched hydrocarbon pro-duction in a strain of Botryococcus braunii. J.Phycol. 21: 388.

Wolf, F. R., Nemethy, E. K., Blanding, J. H., andBassham, J. A. 1985b. Biosynthesis of unusualacyclic isoprenoids in the green alga Botryococcusbraunii. Phytochemistry 24: 733.

Yang, L. W. and Wang, X. Y. 1989. Studies on hydro-carbon production by immobilized Botryococcusbraunii cells. Rev. Roum. Chim. 34: 397.

Yong, T. P. C., Largeau, C., and Casadevall, E. 1986.Biosynthesis of non-isoprenoid hydrocarbons bythe microalga Botryococcus braunii. Evidencesfor elongation- decarboxylation mechanism.Nouv. J. Chim. 10: 701.

Page 35: Botryococcus braunii: A Renewable Source of Hydrocarbons and …ychisti/Botryo.pdf · 2002. 10. 17. · downstream recovery and processing of algal hydrocarbons; the cloning into

279

Zhang, D. and Poulter, C. D. 1995. Biosynthesis ofnon head to tail isoprenoid. Synthesis of 1'-1 and1'-3 structures by recombinant yeast squalenesynthase. J. Am. Chem. Soc. 117: 1641.

Zhang, K. and Kojima, E. 1998. Effect of light inten-sity on colony size of microalga Botryococcusbraunii in bubble column photobioreactor. J.Fermentation Bioeng. 86: 573.