Ribozymes are catalytic RNA molecules that recognize their target RNA in a highly sequence-specific manner. They can therefore be used to inhibit deleterious gene expression (by cleavage of the target mRNA) or even repair mutant cellular RNAs. Targets such as the mRNAs of oncogenes (resulting from base mutations or chromosome translocations, eg, ras or bcr-abl) and viral genomes and transcripts (human immunodeficiency virus–type 1 [HIV-1]) are ideal targets for such sequence-specific agents. The aim of this review is therefore to introduce the different classes of ribozymes, highlighting some of the chemistry of the reactions they catalyze, to address the specific inhibition of genes by ribozymes, the problems yet to be resolved, and how new developments in the field give hope to the future for ribozymes in the therapeutic field.

RIBOZYMES ARE ribonucleic acid (RNA) molecules with enzymatic activity that have a great potential as therapeutic entities because of their ability to either cleave deleterious RNAs or repair mutant cellular RNAs.1,2 They form basepair-specific complexes and catalyze the hydrolysis of specific phosphodiester bonds, causing RNA strand cleavage. Differences exist between ribozymes in size and structure and, although most naturally occurring ribozymes cleave intramolecularly in a cislinkage, the RNA component of RNase-P, which is involved in the processing of pre–t-RNA molecules, acts in trans, ie, intermolecularly.3 

Like its protein counterpart, a catalytic RNA or ribozyme greatly accelerates the rate of a biochemical reaction and shows extraordinary specificity with respect to the substrates it acts upon and the products it produces. Ribozymes can cleave the normally unreactive bonds of a phosphodiester linkage in an RNA molecule resulting in a 3′ hydroxyl (3′OH) and 5′ phosphate (5′OH) and 3′ or 2′3′-cyclic phosphate.4 There are several different classes of ribozymes: the self-splicing group I and group II introns; RNase P; and several distinct catalytic motifs found in the small pathogenic RNAs (Fig 1).

Fig. 1.

RNA catalyzed reactions. (1) The two-step self-splicing reaction of group I introns. (2) The two-step self-splicing reaction of group II introns. An internal hydroxyl initiates the attack. (3) Cleavage of the 5′ leader sequence from pre-tRNA by RNase P. (4) Self-cleavage reaction of a number of small pathogenic RNAs and a few other RNAs.

Fig. 1.

RNA catalyzed reactions. (1) The two-step self-splicing reaction of group I introns. (2) The two-step self-splicing reaction of group II introns. An internal hydroxyl initiates the attack. (3) Cleavage of the 5′ leader sequence from pre-tRNA by RNase P. (4) Self-cleavage reaction of a number of small pathogenic RNAs and a few other RNAs.

Close modal

Many genes have their coding sequences (exons) interrupted by stretches of noncoding DNA called introns. Transcripts of such genes must undergo cleavage-ligation reactions to produce the mature functional RNA. The splicing of most nuclear pre-mRNAs involves a two-step process, generating an intron lariat and spliced exons, and has a requirement for a number of small nuclear ribonucleoprotein particles (snRNPs) and other proteins. Sequences in the RNA components of the snRNPs (U1, U2, and U5 small nuclear RNAs) recognize the 5′ splice site, branch point, and 3′ splice site, respectively, and, together with the U4 and U6 snRNPs, create the spliceosome where the intron is excised and the exons are ligated (Fig 2A; reviewed in Maniatis and Reed5).

Fig. 2.

Comparison of pre-mRNA nuclear splicing, self-splicing, and trans-splicing. (A) Nuclear pre-mRNA splicing with spliceosomes (small nuclear RNAs and proteins). (B) Self-splicing as performed by the self-splicing group I and group II introns. (C)Trans-splicing of a mutated mRNA with a modified group I catalytic intron.

Fig. 2.

Comparison of pre-mRNA nuclear splicing, self-splicing, and trans-splicing. (A) Nuclear pre-mRNA splicing with spliceosomes (small nuclear RNAs and proteins). (B) Self-splicing as performed by the self-splicing group I and group II introns. (C)Trans-splicing of a mutated mRNA with a modified group I catalytic intron.

Close modal

Based on the nucleotide sequences and/or structures within and adjacent to the introns, introns have been classified into four classes: group I, group II, nuclear mRNA, and nuclear tRNA. Some examples of group I and II introns are capable of self-splicing in vitro in the absence of protein (Fig 2B).

Group I introns.

The self-splicing of group I introns, in the presence of a guanosine cofactor and magnesium, was first observed for the intron of the nuclear 26S rRNA gene in Tetrahymena thermophila.6,7 Self-splicing proceeds by two consecutive transesterification reactions, both initiated by nucleophilic attack. The excised intron, with a small deletion, can be converted into a true enzyme able to act in trans on specific substrates.

The RNA cleavage and ligation activities are intrinsic to the structure of the intron. By a number of basepaired regions, both the 3′ and 5′ splice sites are aligned for splicing by the internal guide sequence, close to the guanosine binding site.8-11 The catalytic domain of group I introns is formed by two structural domains, the crystal structures of which are now being determined.12 The interactions between the two domains and nucleotides important for cleavage activity are being elucidated.13 These studies are leading to understanding group I splicing: the mechanism of action and the roles played by the guanosine and metal cofactors.14-16 

Despite not understanding fully the group I intron self-splicing mechanism of action these molecules are being manipulated to performtrans-splicing, ie, the intentional modification of the sequence of a targeted transcript in tissue culture cells.2,17 It can be seen in Fig 2C that a mutation in the RNA sequence can be corrected by replacing part of the mRNA with a new sequence using a suitably modified ribozyme. Although currently not very specific in choice of target RNA (the ribozyme recognizes andtrans-splices several mRNAs), it should be possible to use this approach to develop safe therapeutic ribozymes that can repair mutant RNAs associated with a variety of inherited diseases.

Group II introns.

Some group II introns are also able to undergo self-splicing. They differ from group I introns by the structure of their catalytic core and the products of the splicing: ligated exons and an excised intron-lariat18 (Fig 1). Again, the reaction consists of two transesterifications. The first step can be initiated by nucleophilic attack by an intronic 2′hydroxyl group on the phosphodiester linkage at the 5′ splice site, leading to the formation of the lariat structure. There is evidence for a second pathway, that involving attack by H2O or OH.19The second step is initiated by the 3′OH of the 5′ exon on the 3′ splice site. Basepairing interactions between sequences known as the exon binding site and the intron binding site hold the splice-sites in close proximity.20 This ability of group II introns to specifically bind the 5′ exon has been exploited to encourage the intron to catalyze reactions on exogenous substrates. Derivatives of group II introns reverse splicing by inserting themselves between ligated exons.21 This insertion and subsequent trans-splicing reactions can be used to shuffle sequences22 and therefore link any RNA molecule to any other RNA molecule, intentionally modifying the target sequence.

Ribonuclease P (RNase P) is an ubiquitous endoribonuclease that processes the 5′ end of precursor tRNA molecules, producing 5′ phosphate and 3′OH termini.23 RNase P consists of both protein and RNA components and it was shown that the catalyst was the RNA moiety.24 As with the catalytic introns, a divalent cation is required as cofactor.25 RNase P can be directed to cleave any RNA when the target is in complex with a short, complementary oligonucleotide called an external guide sequence (EGS), thereby inactivating it.26-29 

A number of small plant pathogenic RNAs (viroids, satellite RNAs, and virusoids), an RNA transcript from Neurospora mitochondrial DNA, and an animal virus, HDV, all undergo a self-cleavage reaction in vitro in the absence of protein (Table 1). The reaction, which requires magnesium and produces 2′3′-cyclic phosphate and 5′OH termini, results from several different catalytic motifs: the hammerhead, hairpin, and axehead or pseudoknot (Fig 3). It is generally thought that the self-cleavage reaction is an integral part of the small pathogenic RNAs' rolling circle method of replication.53 Circular monomeric plus and minus RNAs act as templates for the synthesis of longer-than-unit-length precursor RNAs. The production of monomeric forms from these concatamers requires specific cleavage thought to be performed by the RNA itself.

Table 1.

Plant and Animal Pathogenic RNAs and Other RNAs That Self-Cleave In Vitro

References
RNA cleaved by hammerhead structure  
 Avocado sunblotch viroid (ASBV) 30 
 Peach latent mosaic viroid (PLMV) 31,32 
 Potato spindle tuber viroid (PSTV) 33 
 Satellite RNA of Barley yellow dwarf virus (sBYDV) 34 
 Satellite RNA of Tobacco ringspot virus (sTRSV) 35,36 
 Satellite RNA (virusoid) of Lucerne transient streak virus (vLTSV) 37 
 Satellite RNA (virusoid) of Subterranean clover mottle virus (vSCMoV) 38 
 Transcripts from satellite II of the newt 39 
RNA cleaved by hairpin structure  
 sTRSV 40-42 
 Satellite RNA of Chicory yellow mottle virus (sCYMV1) 43 
 Satellite RNA of Arabis mosaic virus (sArMV) 43 
RNA cleaved by other structures (see text)  
 Hepatitis delta virus 44-46 
 Transcripts fromNeurospora mitochondrial DNA plasmid 47-50 
References
RNA cleaved by hammerhead structure  
 Avocado sunblotch viroid (ASBV) 30 
 Peach latent mosaic viroid (PLMV) 31,32 
 Potato spindle tuber viroid (PSTV) 33 
 Satellite RNA of Barley yellow dwarf virus (sBYDV) 34 
 Satellite RNA of Tobacco ringspot virus (sTRSV) 35,36 
 Satellite RNA (virusoid) of Lucerne transient streak virus (vLTSV) 37 
 Satellite RNA (virusoid) of Subterranean clover mottle virus (vSCMoV) 38 
 Transcripts from satellite II of the newt 39 
RNA cleaved by hairpin structure  
 sTRSV 40-42 
 Satellite RNA of Chicory yellow mottle virus (sCYMV1) 43 
 Satellite RNA of Arabis mosaic virus (sArMV) 43 
RNA cleaved by other structures (see text)  
 Hepatitis delta virus 44-46 
 Transcripts fromNeurospora mitochondrial DNA plasmid 47-50 
Fig. 3.

Small catalytic RNAs. (A) Hammerhead ribozyme split into substrate and catalyst,51 basepaired by means of two flanking arms (in bold). Numbering is that of Hertel et al.52 (B) Hairpin ribozyme split into substrate and catalyst.42 (C) Pseudoknot motif of the HDV ribozyme.45 Arrows show position of scissile bonds. IUB codes used throughout: N = A, C, G, U; R = A, G; Y = C, U; B = C, G, U; D = A, U, G; H = A, C, U; V = A, C, G.

Fig. 3.

Small catalytic RNAs. (A) Hammerhead ribozyme split into substrate and catalyst,51 basepaired by means of two flanking arms (in bold). Numbering is that of Hertel et al.52 (B) Hairpin ribozyme split into substrate and catalyst.42 (C) Pseudoknot motif of the HDV ribozyme.45 Arrows show position of scissile bonds. IUB codes used throughout: N = A, C, G, U; R = A, G; Y = C, U; B = C, G, U; D = A, U, G; H = A, C, U; V = A, C, G.

Close modal

The reaction is a simple nonhydrolytic cleavage whereby the scissile bond undergoes a nucleophilic attack by the adjacent 2′OH group (Fig 1).54 The stereochemistry of cleavage and the role of Mg2+ in catalysis has been greatly studied.55,56 In the case of the hammerhead ribozyme, the crystal structure57,58 and fluorescence resonance energy transfer (FRET)59 measurements have shown the relative orientation of the core helices and have allowed the hammerhead catalytic core to be modelled.

Both of the crystallized hammerhead ribozymes contained modifications to prevent self-cleavage: the first57 contained an all DNA substrate analog; the second58, although all RNA, had the active site 2′ hydroxyl replaced with an inert 2′ methoxy group. As such, they may not represent the active structure. Scott et al60 have used time-resolved crystallography to determine the structure of an active hammerhead ribozyme in the absence of divalent cation or with Mg2+ but at a pH unsuitable for cleavage. A captured conformational intermediate showed that the most significant conformational changes were limited to the active site of the ribozyme.

It has been suggested that the function of the ribozyme catalytic core is twofold. First, it would destabilize the substrate strand to allow the scissile phosphodiester linkage to twist into a cleavable conformation. Second, the core would assist in positioning the divalent metal ion so as to facilitate catalysis.57 The more information gained about the active structure of the hammerhead ribozyme, the more rational the design of specific ribozymes can be. Once factors governing the specificity and mechanism of cleavage are elucidated, it may be possible to design new catalysts that efficiently cleave other sequences.

The hammerhead, hairpin, HDV, and Neurospora ribozymes have all been converted from the naturally occurring cis-active ribozymes to trans-active ribozymes by splitting the catalytic core from the substrate sequence. The Neurospora VS ribozyme has been converted once61 and is the least understood motif. More work has been performed on the trans-active HDV ribozyme. The development of this trans-active ribozyme is hampered by the lack of firm knowledge of the secondary and tertiary structure of the RNA and the alternative structures proposed.45,62,63 Recent studies are providing more detailed information.64,65 Despite this lack of understanding of the structure, trans-active HDV ribozymes have been developed.66,67 

The development of the hammerhead and hairpin catalytic domains fortrans-cleavage are far more advanced. The structures and RNA folding are fairly well understood and the models for the design oftrans-acting hammerhead and hairpin ribozymes are well tried and tested.

The hammerhead (Fig 3A) consists of two flanking arms capable of basepairing with the substrate to form two helices (I and III) and a catalytic core with a helix (II) and several single-stranded regions. The ribozyme recognizes sequences either side of a substrate NUH site by means of the flanking arms. The catalytic core then cleaves the RNA 3′ of the NUH triplet. The hairpin ribozyme (Fig 3B) has also been split into substrate and catalytic core. Ribozymes are active in vitro in cell-free systems and in living cells. This activity and their sequence specificity makes them attractive as agents for the inhibition of gene expression.

As described below, they have been used against oncogenes and viruses and have helped in the understanding of the role of genes in developmental processes. Many of the problems with ribozymes are similar to those outlined for antisense oligodeoxy-nucleotides (ODNs).68 Efficient entry into cells, ribozyme stability, and precise targeting to the substrate RNA sequences are receiving attention. Most, if not all, RNAs in vivo normally exist as ribonucleoprotein (RNP) complexes and not as free molecules. Interactions between ribozymes and substrates and these RNP proteins could greatly influence activity. Ribozyme binding could be inhibited by steric hindrance. Alternatively, proteins may enhance activity via annealing and strand-exchange activities. The intracellular environment, such as pH and the availability of divalent cations, must also be considered. For example, the intracellular Mg2+concentration is known to be approximately 0.8 mmol/L,69much lower than is commonly used for in vitro reactions (10 to 20 mmol/L). A primary aim is to show that the ribozyme functions in vivo in a catalytic manner producing two cleavage fragments.

Specificity has, in many cases, been inferred from biological effects without measuring target mRNA or protein levels or looking for ribozyme cleavage products or for effects on unrelated mRNAs. Initial studies of ribozyme activity in cultured cells have come across difficulties in detecting the activity and the catalytic nature of the ribozyme. Several groups have resorted to using a reporter gene, such as chloramphenicol acetyltransferase (CAT),85 in which a reduction in the CAT activity is taken as demonstrating ribozyme activity, or neomycin phosphotransferase (npt).86 Ribozyme specificity has been demonstrated for the βAPP mRNA when this was shown to be reduced compared with control RNAs, α-actin, and G3PDH mRNA.87 However, in this last study, a mutant ribozyme (catalytically inactive) was also effective at reducing levels, suggesting that the ribozyme-mediated degradation of βAPP mRNA in COS-7 cells was not dependent on ribozyme cleavage.

Table 2.

Incomplete List of Potential Specific Gene Inhibition Using Ribozymes

ApplicationRibozyme TargetAffectReferences
Overcoming drug resistance MDR-1 ∼200-fold reduction in vincristine resistance 70 
  Reduction in mRNA 71 
Overcoming transformed H-ras (mutation) Abrogation of tumorigenicity in nude mice 72 
  phenotype  Decrease in proliferation and differentiation in melanoma cells 73 
 bcr-abl (translocation) Decrease in cell proliferation 74 
  Decrease in mRNA and protein levels 75,76 
  Decrease in tumorigenicity ofbcr-abl–transfected cells in nude mice 77 
 Pleiotrophin (growth factor) Decrease in tumour growth, angiogenesis and metastasis 78 
Overcoming viral disease HIV-1 Increased resistance to HIV-1 infection 79 
  CD4(+) lymphocytes transduced with ribozymes has delayed reaction to viral challenge 80 
  Inhibition of HIV-1 replication 81 
 Hepatitis B and C virus  82,83 
Overcoming arthritis Stromelysin mRNA reduced 84 
ApplicationRibozyme TargetAffectReferences
Overcoming drug resistance MDR-1 ∼200-fold reduction in vincristine resistance 70 
  Reduction in mRNA 71 
Overcoming transformed H-ras (mutation) Abrogation of tumorigenicity in nude mice 72 
  phenotype  Decrease in proliferation and differentiation in melanoma cells 73 
 bcr-abl (translocation) Decrease in cell proliferation 74 
  Decrease in mRNA and protein levels 75,76 
  Decrease in tumorigenicity ofbcr-abl–transfected cells in nude mice 77 
 Pleiotrophin (growth factor) Decrease in tumour growth, angiogenesis and metastasis 78 
Overcoming viral disease HIV-1 Increased resistance to HIV-1 infection 79 
  CD4(+) lymphocytes transduced with ribozymes has delayed reaction to viral challenge 80 
  Inhibition of HIV-1 replication 81 
 Hepatitis B and C virus  82,83 
Overcoming arthritis Stromelysin mRNA reduced 84 

Ribozyme cleavage has been demonstrated by an RNase protection assay, where the cleavage products protected the probe.86 But care has to be taken in interpreting such results. It was recently noted that a hammerhead ribozyme cleaved its substrate during RNA preparation and not while in the cell.82,88 

Despite some of the problems already alluded to, many groups are actively investigating ribozymes' therapeutic applications.

The reversal of drug resistance is a popular goal for ribozyme technology. Kobayashi et al70 showed they could reduce MOLT-3 cells from approximately 700-fold resistant to vincristine to only 20- to 30-fold resistant when an anti–MDR-1 ribozyme was transfected in and stably expressed (under the β-actin promoter). This increase in drug sensitivity, and reduction in MDR-1 mRNA levels, was shown to be proportional to the amount of ribozyme expression. They critically showed that, although they could not detect cleavage products, a disabled ribozyme incapable of cleavage had no effects on the drug resistance levels. They also tested their ribozyme in the non–drug-resistant parental cell line and found no effect: the ribozyme was not toxic. Kiehntopf et al71 found that liposome-mediated transfection of drug-resistant mesothelioma cell lines with either in vitro transcribed or chemically synthesized ribozymes significantly reduced expression of the MDR-1 gene. This restored sensitivity towards chemotherapeutic drugs. They were also able to demonstrate, using reverse transcription polymerase chain reaction, the reduction in full-length MDR-1 mRNA levels.

The mRNAs from oncogenes have also been targeted by ribozyme technology. H-ras is activated by a mutation at codon 12 (GGU → GUU), with the activated form being a substrate for hammerhead ribozyme cleavage. When transformed NIH3T3 cells that displayed a neoplastic phenotype in vitro and were tumorigenic in nude mice in vivo were transfected by a ribozyme designed to cleave the activated H-ras mRNA, the transformed phenotype was abrogated.72 A reduction in H-rasmRNA was observed. A mutant ribozyme resulted in cells with an intermediate phenotype, probably due to an antisense effect of the ribozyme hybridizing arms. This group also addressed the question of specificity by looking at the K-ras mRNA levels in control and ribozyme-treated cells and showed no cross-reactivity. Ohta et al73 demonstrated that a tissue-specific promoter for the expression of an anti–H-ras ribozyme produced better results than using a viral promoter. This result suggests that, depending on the target mRNA, the choice of viral promoter for ribozyme expression is not always the best option. The transfected ribozyme appeared to affect not only proliferation but also the differentiation process of the melanoma cells in vitro.89 

BCR-ABL in chronic myeloid leukemia.

Chromosome translocations and the resulting chimaeric genes are good targets for sequence-specific strategies. The hybrid mRNA will only be present in the cells with the translocation, and antisense ODNs or ribozymes targeted to the hybrid's junction should be specific for the hybrid and not affect the wild-type mRNA sequences. One such translocation results in the Philadelphia chromosome (Ph+) of chronic myeloid leukemia, and the bcr-abl oncogene. The expression of the bcr-abl protein tyrosine kinase is thought to be responsible for the malignant phenotype of Ph+ cells. Both the wild-type abl and bcr proteins are thought to be important for normal cell proliferation. This makes Ph+cells the ideal system to address the question of ribozyme sequence specificity. We have looked at the specificity of three different hammerhead ribozymes designed to cleave two splice variants of thebcr-abl mRNA90 in an attempt to clear up some of the contradictory results published.91-93 We showed the specific nature of one ribozyme (cleaved only bcr-abl RNA) but a lack of specificity of two others (cleaved both bcr-abl andabl RNAs). In a cell line system, the question of specificity has not really been addressed, but a decrease in cell proliferation,74 a decrease in bcr-abl mRNA and protein levels,75,76 and a decrease in bcr-ablkinase activity93 have been observed (Table 3). These studies all used slightly different hammerhead ribozymes: varying lengths of the flanking arms and various modifications to improve nuclease resistance or expression from vectors. Some groups observed quite substantial effects,75,76,93 whereas others have seen more modest91 or shown very few effects (our unpublished data). This is probably due to differences in the ribozymes' structures. Until the structure-function relationship is understood, such effects will not be predictable.

Table 3.

Effects of bcr-abl Ribozymes in Cell Lines

ReferenceCell LineTransfection MethodRobozymeEffects
74,91 K562 DOTAP Unmodified RNA Uptake followed by fluorescence, ribozyme stable up to 12 hours Decrease in proliferation 
75 EM-2, HL60 Transfectam  DNA-RNA hybrid Decrease inbcr-abl mRNA, protein expression, growth 
   (lipopolyamine)  No observable effects on p160bcr levels, β-actin mRNA unaffected Unrelated ribozyme had very limited effect 
93 K562 Various vectors  Decrease inbcr-abl kinase activity, cell number 
    Vector and ribozyme expression level dependent (RNA polymerase III tRNA vector best) 
76 32Dbcr-abl Lipofectin Folic acid-poly lysine  Unmodified RNA Slight effect on mRNA levels Decrease in bcr-abl mRNA levels Inactive ribozyme had no effect on mRNA levels 
77 32Db3a2 in SCID mice Lipofectamine DNA-RNA hybrid Increase in survival of SCID mice after injection with ribozyme treated 32Db3a2 cells 
ReferenceCell LineTransfection MethodRobozymeEffects
74,91 K562 DOTAP Unmodified RNA Uptake followed by fluorescence, ribozyme stable up to 12 hours Decrease in proliferation 
75 EM-2, HL60 Transfectam  DNA-RNA hybrid Decrease inbcr-abl mRNA, protein expression, growth 
   (lipopolyamine)  No observable effects on p160bcr levels, β-actin mRNA unaffected Unrelated ribozyme had very limited effect 
93 K562 Various vectors  Decrease inbcr-abl kinase activity, cell number 
    Vector and ribozyme expression level dependent (RNA polymerase III tRNA vector best) 
76 32Dbcr-abl Lipofectin Folic acid-poly lysine  Unmodified RNA Slight effect on mRNA levels Decrease in bcr-abl mRNA levels Inactive ribozyme had no effect on mRNA levels 
77 32Db3a2 in SCID mice Lipofectamine DNA-RNA hybrid Increase in survival of SCID mice after injection with ribozyme treated 32Db3a2 cells 

The hybrid gene AML1/MTG8 mRNA that results from a translocation between chromosomes 8 and 21 (associated with acute myeloid leukemia) is also being targeted by hammerhead ribozymes.94 

Another clinical situation amenable to ribozyme targeting is viral disease, such as that associated with the human immunodeficiency virus (HIV). Two classes of ribozymes are being used to combat HIV: hammerhead79,81,95,96 and hairpin.97 

Weerasinghe et al95 showed that MT4 cells transformed with vectors expressing an HIV-1 ribozyme were resistant to varying degrees of HIV-1 infection. The choice of constitutive versus inducible promoters for the expression of the anti–HIV-1 ribozymes had quite an influence on the degree of resistance to HIV-1 infection. A similar study was performed by Lo et al96 with a hammerhead ribozyme designed to cleave the HIV-1 tat RNA. Interestingly, although the anti-tat ribozyme-producing cells inhibited the replication of HIV-1, an antisense producing vector conferred a greater resistance to HIV-1 replication.

A third group have shown that T lymphocytes expressing anti–HIV-1 ribozymes showed resistance to HIV-1 replication.79 Cells transformed with a mutant ribozyme showed little resistance to viral replication. They argued that this demonstrated that the functional ribozymes were cleaving the HIV-1 RNA and were specific for this RNA. They have also looked at the effects of using different retroviral vectors to express the ribozymes: despite different ribozyme levels, there was a similar level of resistance to HIV-1 replication.98 This indicated that other factors had determining roles in the effectiveness of the ribozyme.

Bauer et al81 have made an important step by treating CD34(+) cells from individuals already infected with HIV-1. They transfected the cells with a retroviral vector containing a doubletat-rev ribozyme and showed up to a 1,000-fold inhibition of HIV-1 replication after further challenge with the virus.

A hairpin ribozyme has been shown to have similar effects to the hammerhead ribozyme: cells expressing the ribozyme were resistant to various strains of HIV-1. It was also demonstrated that the hairpin ribozyme significantly reduced the efficiency of the incoming virus to synthesize viral DNA.97 A hairpin ribozyme is in the preliminary stages of a phase I clinical trial against HIV-1.99 CD4(+) lymphocytes from HIV-1–infected donors were transduced with ribozyme or control vectors and viral replication was delayed by 2 to 3 weeks.80 The rapid progress of ribozymes to early clinical trials can be attributed to their specificity and ability to be delivered by viral vectors to the target cells. Another reason ribozymes could be successful against retroviruses is that they can potentially target several stages in the viral life cycle: the incoming genomic RNA, the mRNA transcribed from the integrated genome, and the progeny RNA genomes.

HIV-1 is not the only virus being targeted. Beck and Nassal82 are targeting a hammerhead ribozyme to the encapsidation signal ε of Hepatitis-B virus, the causative agent of B-type hepatitis. Unfortunately, ribozyme cleavage has only been observed in vitro and in cell extracts, but not in intact cells. Even when the ribozyme was placed in cis with an artificial substrate, little ribozyme activity was observed (most of the activity could be attributed to antisense effects). The investigators speculated that there was something inhibiting cleavage within the cell, because efficient cleavage was observed upon RNA extraction. Hepatitis-C virus RNA is also the subject of selective targeting and destruction by ribozymes,83,100,101 as is the mRNA for influenza A virus.102 

Some researchers are beginning to assay ribozyme effects in animal model systems.77,84,103 We have been investigating the action of an ex vivo purging of a murine cell line containing thebcr-abl oncogene with ribozymes against the bcr-ablmRNA before injection into SCID mice. Effectiveness of the ribozyme is demonstrated by the increased survival of the mice (when compared with mice injected with control-treated bcr-abl–containing cells).77 

Flory et al84 have used a rabbit model of interleukin-1–induced arthritis to assess the localization, stability, and efficacy of exogenous anti-stromelysin hammerhead ribozymes. They observed that exogenously delivered ribozymes were taken up by cells in the synovial lining and synovial interleukin-1α–induced stromelysin mRNA levels were reduced. Catalytically inactive ribozymes were ineffective supporting a cleavage mechanism for ribozyme activity. Using nude mice, Czubayko et al78 have rather nicely demonstrated the relationship between the secreted growth factor pleiotrophin (PTN) from melanoma cells and the melanoma cells' metastasis to the lungs. Introducing ribozymes against PTN into a cell line, which reduced PTN mRNA and growth factor activity, concomitantly prevented the metastatic spread of the tumors. Larsson et al103 looked at hammerhead ribozymes against the β2-microglobulin (β2M) mRNA in both cell lines and transgenic mice. They observed ribozyme expression in lung, kidney and spleen with greatest reduction in the β2M mRNA levels in the lung. However, it should be noted that ribozyme levels and reduction in β2M mRNA varied widely between litter mates. Lieber and Kay104 have looked at ribozymes (against human growth hormone) expressed from adenovirus vectors after somatic gene transfer into transgenic mice. A reduction of up to 96% in growth hormone was observed in correlation with the mRNA levels. Using transgenic mice is obviously quite different from using a retroviral vector to express a ribozyme. Although inappropriate for therapeutic administration, transgenic animals can provide important information about ribozyme expression levels in different tissues and ribozyme efficacy, as well as information about the role of the targeted mRNA.

Very little has been done so far with respect to pharmacokinetic and pharmacodynamic data for ribozymes in animal model systems. However, one study of pharmacokinetic properties of synthetic, chemically modified hammerhead ribozymes after intravenous injections105 has shown the prolonged presence of a cytochrome P-450 ribozyme in the plasma (up to 48 hours postinjection) and perfusion into tissues other than the vascular system (kidney and liver). This group has also injected a similarly modified ribozyme to amelogenin mRNA, with no carrier to assist cellular uptake, and shown a surprisingly successful knock-out of the targeted gene's expression.106 The various works detailing transgenic animals with a gene construct for an antisense RNA or ribozyme sequence has recently been reviewed.107 

As well as their potential as therapeutic agents, ribozymes can be used to generate loss-of-function phenotypes to elucidate the roles of genes. This has been performed for the Fushi taragu gene in Drosophila,108 for c-fos,109 and for matrix metalloproteinase 2.110 

Ribozymes have also been used in other systems to varying degrees of success: in plant protoplasts111,112 and in yeast (Saccharomyces cerevisiae113 and S pombe114). Transgenic plants with ribozymes to the lignin-forming peroxidase of tobacco have been created, with reduction in the peroxidase mRNA and protein levels observed.115 

(For assistance in designing and applying ribozyme technology please refer to two recently published methods books: Ribozyme Protocols116 and Antisense and Ribozyme Methodology.117)

As detailed above, ribozymes have been successfully used to target specific mRNAs. To use a ribozyme strategy, several parameters must be decided. The first choice is the type of ribozyme to use. The hammerhead ribozyme is the best-studied type. The target sequence requirements for the hammerhead are slightly less stringent than for the hairpin, and, because the hammerhead is smaller than the hairpin, it is cheaper to synthesize and higher yields can be obtained. The next decision is the choice of target mRNA. It has been suggested that targeting the mutated gene sequence may not always be the most effective approach. Targeting an mRNA of a protein downstream of the mutated gene product may result in a more effective inhibition. However, this could lead to a decrease in specificity between normal and affected cells. The position of the NUH target site within the mRNA must then be selected. The efficiency of the ribozyme is dictated by the sequence (different NUH sequences can vary by >100-fold in the rates of their cleavage, and the sequence context of the site is also an important factor118) and accessibility of target site due to secondary and tertiary structures of the mRNA.119Obviously, before cleavage can occur, the ribozyme must bind to its substrate via the sequences either side of the target site. Experimental analysis such as nuclease mapping or chemical probing can be performed to monitor accessibility.120 Alternatively computer RNA folding programs can be used to help determine accessible sites121,122 with less expense but at a reduced reliability.

Most computational algorithms for RNA structure prediction are based on calculation of the minimum free energy (ΔG). For a stretch of bases the program determines the most stable structure, the lowest ΔG. The computer then looks at the next stretch of bases (which may or may not overlap with the previous stretch) and calculate the next ΔG. In this way, local maxima and minima ΔG values are obtained that correspond to relatively unstable or stable regions of the RNA, respectively. The value of ΔG is dependent on which algorithm is used to determine the structure, and this is where the errors can be introduced. A minima in ΔG does not necessarily mean that a stable structure exists in vitro or in vivo. Current programs are also limited to the length of the sequence to be analyzed; distant interactions in a long mRNA cannot be predicted.

The length of the ribozyme's hybridizing arms (which basepair with the substrate) must strike a balance between providing specificity for the substrate while allowing product dissociation.123,124 It has been suggested that asymmetric arms may be more effective than symmetric arms.125 It is also important to realize that results suggesting an efficient arm length in a cell-free system may not be ideal in a cellular system.126,127 An increasing number of laboratories are applying in vitro selection, or in vitro evolution, techniques (SELEX) to develop more efficient ribozymes and ribozymes with new activities.128,129 The principal of the technique is based on the screening by selection of a pool of RNA molecules. Starting with either a totally or partially random pool of RNA molecules, a cycling of binding or activity, partitioning, and amplification steps results in a novel ribozyme or ribozymes with the desired binding or catalytic activity. Using this technique, and by chance, both RNA and antisense DNA molecules have been developed that are aptameric in activity, ie, they can interact directly with the target protein in a sequence-specific manner.130-132 

Many groups are looking at ways to improve ribozyme catalytic activity. One way to do this is to use multiunit ribozymes (ie, transcripts with more than one catalytic domain) targeting different sites within the same mRNA. Chen et al133 developed a nona-ribozyme against HIV-1 and showed that this was more effective at cleaving substrate RNA in vitro and inhibiting HIV-1 replication in cell lines than mono-ribozymes. This multimeric ribozyme has a second advantage, ie, HIV-1 escape mutants are less likely. A multiunit anti–BCR-ABLribozyme (3 catalytic domains) was shown to be more effective than the three ribozyme domains added separately, both in vitro and in cell lines, by reducing the BCR/ABL mRNA.134 

Efforts to increase exogenously delivered ribozymes' stability and hence activity have concentrated on chemical modifications to the ribozyme structure. 2′-fluoro or 2′-amino135and 2′-O-allyl and 2′-O-methyl106,136 are a few of the modifications being investigated.

Ribozymes can be delivered to cells in two ways: as preformed ribozymes (exogenous delivery) or as ribozyme genes, a method of endogenous delivery. The means of transfection (eg, lipofection or electroporation) is important for the former, whereas the choice of promoter (pol II, pol III, viral, etc) is important for the latter.137 The latter could be termed ribozyme gene therapy, whereby the introduced ribozymes downregulate the targeted gene expression or repair mutant mRNAs.138 

Endogenous delivery has been achieved by inserting ribozyme sequences into the untranslated regions (UTRs) of genes transcribed by RNA polymerase II (pol II), such as the SV40 early promoter85or the actin gene.139 The RNA polymerase III (pol III) promoters from the U6 small nuclear RNA140 or from certain tRNAs have also been successfully used to express ribozymes and achieve effects.141 Tissue-specific promoters, such as the tyrosinase promoter, have also been investigated.73 

Both retroviral-derived and more recently adeno-associated viral (AAV)-derived vectors are commonly used. Ribozyme genes can be expressed from the viral long terminal repeat (LTR) promoters or from introduced pol II or pol III promoters. Retroviral vectors are relatively efficient, safe, and capable of (randomly) integrating stably in the host genome of replicating cells. AAV is nonpathogenic and has the advantage of integrating into a defined region of the host genome without requiring cell division. Zhou et al98compared three types of promoters: Molony murine leukemia virus (MoMuLV) LTR, the human CMV promoter, and a human tRNAmetcassette for the transcriptional control of a pair of anti–HIV-1 ribozymes. The LTR promoter produced the highest expression levels of the ribozyme, but the ability of each to confer resistance to HIV-1 replication was very similar. The investigators concluded that other factors (antisense effects, protein influence, and localization) than the absolute levels of the ribozymes played a role in determining the effectiveness of the ribozymes. The recent work of Bertrand et al137 shows that colocalization is the most important factor. In this study, promoters used for the expression cassettes were derived from the human tRNAmeti (pol III), the human U1 snRNA (pol II), the human U6 snRNA (pol III), and the Rous sarcoma virus (RSV) LTR (pol II). Each cassette was introduced into a cell line with both AAV- and MoMuLV-based vectors. All of the cassettes produced ribozymes when in context of the AAV vector, whereas the tRNA and U6 cassettes were inactive with the viral vector. Anti-HIV ribozymes derived from the RSV cassette were expressed at the lowest levels and were cytoplasmic (consistant with being capped and polyadenylated). The other transcripts were predominantly nuclear and expressed at higher levels. When the cells were challenged with HIV, surprisingly only those containing the capped, poly-A RNAs that were cytoplasmically localized were able to suppress HIV replication.

Sullenger and Cech142 have successfully looked at using a viral packaging signal to direct their ribozymes to the same subcellular locations as the viral targets. It has also been observed that the 3′UTRs of some mRNAs carry the signal responsible for localization.143,144 These, and others yet to be discovered, could be attached to ribozyme sequences to aid their colocalization. In a hope to understand RNA trafficking in the cell, we have been following ribozyme distribution using fluorescently tagged ribozymes.145 Because the nucleus is thought by many to be the site of action for ribozymes, several groups are looking at ribozyme activity when expressed with a nuclear localization signal140 or in isolated nuclei.146 It can be concluded that the type of promoter and its context can determine the intracellular compartmentalization of the ribozyme and that colocalizing the ribozyme with the substrate is a primary determinant of ribozyme efficacy in vivo.

Measurements for assaying ribozyme activity can be made at the mRNA (RNase protection assays or reverse-ligation polymerase chain reaction147) or protein levels (Western blotting and antibody probing), function of the protein (eg, kinase activity) and effects on cell differentiation, or the onset of apoptosis. In general, ribozyme cleavage activity in the cell system or in vivo has not been empirically demonstrated, but instead has been inferred from the lack of effects by a catalytically inactive ribozyme if included as a control.

Stein and Krieg148 pointed out various considerations for the interpretation of data derived from the use of antisense ODNs. Charged ODNs are polyanions and as such can bind and sequester growth factors to the basement membrane. Phosphorothioate ODNs may exhibit nonantisense but sequence-dependent effects, eg, a G quartet149 or a TAT triplet at the 3′ end.150 These considerations, although not proven, are probably just as valid for ribozymes. To show specific ribozyme cleavage, the following controls are required: a catalytically inactive ribozyme (to show antisense effects), a conventional antisense ODN equivalent to the ribozyme arms (again, to show antisense effects), an active but unrelated ribozyme (to show specificity for target mRNA), a scrambled ODN, and finally the transfection agent or vector only (nonspecific toxicity effects).

Ribozymes and antibiotics.

It has been demonstrated that certain classes of antibiotics can interact with some ribozymes and, in some cases, influence their cleavage activity. Aminoglycoside antibiotics inhibit group I intron function but not group II introns.151 The aminoglycosides, in particular neomycin B, have been shown to inhibit hammerhead ribozyme activity,152 as have the tetracyclins.153 The HDV ribozyme is also inhibited by both aminoglycosides and tetracyclins.154 Contrary to this, theNeurospora VS ribozyme cleavage activity is actually enhanced by viomycin, a tuberactinomycin antibiotic.155 The modulation of ribozyme activity by other agents, in particular antibiotics, should be taken into consideration when designing experiments with cell cultures (often grown in the presence of antibiotics) and possibly in a clinical setting.

For ribozymes to become realistic therapeutic agents several obstacles need first to be overcome. These obstacles are the efficient delivery to a high percentage of the cell population, efficient expression of the ribozyme from a vector or intracellular ribozyme concentration, colocalization of the ribozyme with the target, specificity of ribozyme for the desired mRNA, and an enhancement of ribozyme-mediated substrate turnover.

Despite these reservations, results with ribozymes so far look promising, particularly in the HIV-1 studies. As our knowledge of RNA structure, secondary and tertiary, increases, we will be able to target the RNA more rationally, which may help with the problems of specificity. At the same time, the understanding of the physical localization of RNA in cells and its tracking as it moves from the nucleus to cytoplasm will also help in ensuring colocalization of the ribozyme and target. Modifications of the ribozymes, eg, the 2′ ribose with allyl group, increases the stability to nucleases quite dramatically. Similarly, DNA sequences allied to the ribozymes in use in our laboratory increase the stability. Entry into cells with liposomes or via vectors are also looking hopeful. Catalytic activity is maintained but research is still ongoing at tackling the problem of increasing the number of ribozyme molecules in proportion to the substrate molecules—a requirement for success. These molecules must retain their catalytic potential, must reach an accessible site in the substrate, and eventually be synthesized from the appropriate vector chosen for clinical trials. Work in the antisense DNA field would also benefit from solutions to these problems.

Supported by Grant No. 9667 of the Leukaemia Research Fund, London, UK.

Address reprint requests to Helen A. James, PhD, School of Biological Sciences, University of East Anglia, Norwich, Norfolk, NR4 7TJ UK.

1
Rossi
JJ
Cantin
EM
Sarver
N
Chang
PF
The potential use of catalytic RNAs in therapy of HIV infection and other diseases.
Pharmacol Ther
50
1991
245
2
Sullenger
BA
Cech
TR
Ribozyme-mediated repair of defective mRNA by targeted trans-splicing.
Nature
371
1994
619
3
James
HA
Turner
PC
Ribozymes.
Essays Biochem
29
1995
175
4
Cedergren
R
RNA—The catalyst.
Biochem Cell Biol
68
1990
903
5
Maniatis
T
Reed
R
The role of small nuclear ribonucleoprotein particles in pre-mRNA splicing.
Nature
325
1987
673
6
Cech
TR
Zaug
AJ
Grabowski
PJ
In vitro splicing of the ribosomal RNA precursor of Tetrahymena: Involvment of a guanosine nucleotide in the excision of the intervening sequence.
Cell
27
1981
487
7
Kruger
K
Grabowski
PJ
Zaug
AJ
Sands
J
Gottschling
DE
Cech
TR
Self-splicing RNA: Autoexcision and autocyclization of the ribosomal RNA intervening sequence of Tetrahymena.
Cell
31
1982
147
8
Been
MD
Cech
TR
One binding site determines sequence specificity of Tetrahymena pre-rRNA self-splicing, trans-splicing and RNA enzyme activity.
Cell
47
1986
207
9
Garriga
G
Lambowitz
AM
Inoue
T
Cech
TR
Mechanism of recognition of the 5′ splice site in self-splicing group I introns.
Nature
322
1986
86
10
Inoue
T
Sullivan
FX
Cech
TR
New reactions of the ribosomal RNA precursor of Tetrahymena and the mechanism of self-splicing.
J Mol Biol
189
1986
143
11
Suh
ER
Waring
RB
Base pairing between the 3′ exon and an internal guide sequence increases 3′ splice site specificity in the Tetrahymena self-splicing rRNA intron.
Mol Cell Biol
10
1990
2960
12
Cate
JH
Gooding
AR
Podell
E
Zhou
K
Golden
BL
Kundrot
CE
Cech
TR
Doudna
JA
Crystal structure of a group I ribozyme domain: principles of RNA packing.
Science
273
1996
1678
13
Tanner
MA
Cech
TR
Joining the two domains of a group I ribozyme to form the catalytic core.
Science
275
1997
847
14
Golden
BL
Cech
TR
Conformational switches involved in orchestrating the successive steps of group I RNA splicing.
Biochemistry
35
1996
3754
15
Mei
R
Herschlag
D
Mechanistic investigations of a ribozyme derived from the Tetrahymena group I intron—Insights into catalysis and the second step of self-splicing.
Biochemistry
35
1996
5796
16
Sjogren
AS
Pettersson
E
Sjoberg
BM
Stromberg
R
Metal ion interaction with cosubstrate in self-splicing of group I introns.
Nucleic Acids Res
25
1997
648
17
Jones
JT
Lee
S-W
Sullenger
BA
Tagging ribozyme reaction sites to follow trans-splicing in mammalian cells.
Nat Med
2
1996
643
18
Peebles
CL
Perlman
PS
Mecklenburg
KL
Petrillo
ML
Yabor
JH
Jarrell
KA
Cheng
H-L
A self-splicing RNA excises an intron lariat.
Cell
44
1986
213
19
Daniels
DL
Michels
WJ
Jr
Pyle
AM
Two competing pathways for self-splicing by group II introns: A quantitative analysis of in vitro reaction rates and products.
J Mol Biol
256
1996
31
20
Jacquier
A
Rosbash
M
Efficient trans-splicing of a yeast mitochondrial RNA group II intron implicates a strong 5′ exon-intron interaction.
Science
234
1986
1099
21
Morl
M
Schmelzer
C
Integration of group II intron bl1 into a foreign RNA by reversal of the self-splicing reaction in vitro.
Cell
60
1990
629
22
Mikheeva
S
Jarrell
KA
Use of engineered ribozymes to catalyze chimeric gene assembly.
Proc Natl Acad Sci USA
93
1996
7486
23
Darr
SC
Brown
JW
Pace
NR
The varieties of ribonuclease P.
Trends Biol Sci
17
1992
178
24
Guerrier-Takada
C
Gardiner
K
Marsh
T
Pace
N
Altman
S
The RNA moiety of ribonuclease P is the catalytic subunit of the enzyme.
Cell
35
1983
849
25
Guerrier-Takada
C
Haydock
K
Allen
L
Altman
S
Metal ion requirements and other aspects of the reaction catalyzed by M1 RNA, the RNA subunit of ribonucleases P from Escherichia coli.
Biochemistry
25
1986
1509
26
Yuan
Y
Altman
S
Selection of guide sequences that direct efficient cleavage of mRNA by human ribonuclease P.
Science
263
1994
1269
27
George ST, Werner M, Nordstrom JL, Goldberg AR: Derivation of a small external guide sequence capable of inducing cleavage of target RNA by human RNase P. J Cell Biochem S19A:209, 1995 (abstr)
28
Werner M, Iannuccilli WJ, George ST, Ma MYX, Goldberg AR: Characterization of the external guide sequence directed cleavage reaction of target RNA molecules with ribonuclease P. J Cell Biochem S19A:213, 1995 (abstr)
29
Li
Y
Altman
S
Cleavage by RNase P of gene N mRNA reduces bacteriophage l burst size.
Nucleic Acids Res
24
1996
835
30
Hutchins
CJ
Rathjen
PD
Forster
AC
Symons
RH
Self-cleavage of plus and minus RNA transcripts of avocado sunblotch viroid.
Nucleic Acids Res
14
1986
3627
31
Hernandez
C
Flores
R
Plus and minus RNAs of peach latent mosaic viroid self-cleave in vitro via hammerhead structures.
Proc Natl Acad Sci USA
89
1992
3711
32
Beaudry
D
Bussiere
F
Lareau
F
Lessard
C
Perreault
J-P
The RNA of both polarities of the peach latent mosaic viroid self-cleaves in vitro by single hammerhead structures.
Nucleic Acids Res
23
1995
745
33
Robertson
HD
Rosen
DL
Branch
AD
Cell-free synthesis and processing of an infectious dimeric transcript of potato spindle tuber viroid RNA.
Virology
142
1985
441
34
Miller
WA
Hercus
T
Waterhouse
PM
Gerlach
WL
A satellite RNA of barley yellow dwarf virus contains a novel hammerhead structure in the self-cleavage domain.
Virology
183
1991
711
35
Buzayan
JM
Gerlach
WL
Bruening
G
Non-enzymatic cleavage and ligation of RNAs complementary to a plant virus satellite RNA.
Nature
323
1986
349
36
Prody
GA
Bakos
JT
Buzayan
JM
Schneider
IR
Bruening
G
Autolytic processing of dimeric plant virus satellite RNA.
Science
231
1986
1577
37
Forster
AC
Symons
RH
Self-cleavage of plus and minus RNAs of a virusoid and a structural model for the active sites.
Cell
49
1987
211
38
Davies
C
Haseloff
J
Symons
RH
Structure, self-cleavage and replication of two viroid-like satellite RNAs (virusoids) of subterranean clover mottle virus.
Virology
177
1990
216
39
Epstein
LM
Gall
JG
Self-cleaving transcripts of satellite DNA from the newt.
Cell
48
1987
535
40
Hampel
A
Tritz
R
RNA catalytic properties of the minimum (-) sTRSV sequence.
Biochemistry
28
1989
4929
41
Chowrira
BM
Burke
JM
Binding and cleavage of nucleic acids by the ‘hairpin’ ribozyme.
Biochemistry
30
1991
8518
42
Berzal-Herranz
A
Joseph
S
Chowrira
BM
Butcher
SE
Burke
JM
Essential nucleotide sequences and secondary structure elements of the hairpin ribozyme.
EMBO J
12
1993
2567
43
DeYoung
MB
Siwkowski
AM
Lian
Y
Hampel
A
Catalytic properties of hairpin ribozymes derived from chicory yellow mottle virus and arabis mosaic virus satellite RNAs.
Biochemistry
34
1995
15785
44
Wu
H-N
Lin
Y-J
Lin
F-P
Makino
S
Chang
M-F
Lai
MMC
Human hepatitis δ virus RNA subfragments contain an autocleavage activity.
Proc Natl Acad Sci USA
86
1989
1831
45
Perrotta
AT
Been
MD
A pseudoknot-like structure required for efficient self-cleavage of hepatitis delta virus RNA.
Nature
350
1991
434
46
Wu
H-N
Wang
Y-J
Hung
C-F
Lee
H-J
Lai
MMC
Sequence and structure of the catalytic RNA of hepatitis delta virus genomic RNA.
J Mol Biol
223
1992
233
47
Saville
BJ
Collins
RA
A site-specific self-cleavage reaction performed by a novel RNA in Neurospora mitochondria.
Cell
61
1990
685
48
Collins
RA
Olive
JE
Reaction conditions and kinetics of self-cleavage of a ribozyme derived from Neurospora VS RNA.
Biochemistry
32
1993
2795
49
Beattie
TL
Olive
JE
Collins
RA
A secondary-structure model for the self-cleaving region of Neurospora VS RNA.
Proc Natl Acad Sci USA
92
1995
4686
50
Rastogi
T
Beattie
TL
Olive
JE
Collins
RA
A long-range pseudoknot is required for activity of the Neurospora VS ribozyme.
EMBO J
15
1996
2820
51
Haseloff
J
Gerlach
WL
Simple RNA enzymes with new and highly specific endoribonuclease activities.
Nature
334
1988
585
52
Hertel
KJ
Pardi
A
Uhlenbeck
OC
Koizumi
M
Ohtsuka
E
Uesugi
S
Cedergren
R
Eckstein
F
Gerlach
WL
Hodgson
R
Symons
RH
Numbering system for the hammerhead.
Nucleic Acids Res
20
1992
3252
53
Branch
AD
Robertson
HD
A replication cycle for viroids and other small infectious RNA's.
Science
223
1984
450
54
Sigurdsson
ST
Eckstein
F
Structure-function relationships of hammerhead ribozymes: From understanding to applications.
Trends Biotech
13
1995
286
55
Dahm
SC
Uhlenbeck
OC
Role of divalent metal ions in the hammerhead RNA cleavage reaction.
Biochemistry
30
1991
9464
56
Yarus M: How many catalytic RNAs? Ions and the Cheshire cat conjecture. FASEB J 7:31, 1993
57
Pley
HW
Flaherty
KM
McKay
DB
Three-dimensional structure of a hammerhead ribozyme.
Nature
372
1994
68
58
Scott
WG
Finch
JT
Klug
A
The crystal structure of an all-RNA hammerhead ribozyme: A proposed mechanism for RNA catalytic cleavage.
Cell
81
1995
991
59
Tuschl
T
Gohlke
C
Jovin
TM
Westhof
E
Eckstein
F
A three-dimensional model for the hammerhead ribozyme based on fluorescence measurements.
Science
266
1994
785
60
Scott
WG
Murray
JB
Arnold
JRP
Stoddard
BL
Klug
A
Capturing the structure of a catalytic RNA intermediate: The hammerhead ribozyme.
Science
274
1996
2065
61
Guo
HCT
Collins
RA
Efficient trans-cleavage of a stem-loop RNA substrate by a ribozyme derived from Neurospora VS RNA.
EMBO J
14
1995
368
62
Branch
AD
Robertson
HD
Efficient trans cleavage and a common structural motif for the ribozymes of the human hepatitis δ agent.
Proc Natl Acad Sci USA
88
1991
10163
63
Gottlieb
PA
Prasad
Y
Smith
JB
Williams
AP
Dinter-Gottlieb
G
Evidence that alternate foldings of the hepatitis δ RNA confer varying rates of self-cleavage.
Biochemistry
33
1994
2802
64
Branch
AD
Polaskova
JA
3-D models of the antigenomic ribozyme of the hepatitis delta agent with eight new contacts suggested by sequence-analysis of 188 cDNA clones.
Nucleic Acids Res
23
1995
4180
65
Jeng
K-S
Daniel
A
Lai
MMC
A pseudoknot ribozyme structure is active in vivo and required for hepatitis delta virus RNA replication.
J Virol
70
1996
2403
66
Been
MD
Cis- and trans-acting ribozymes from a human pathogen, hepatitis delta virus.
Trends Biol Sci
19
1994
251
67
Kawakami
J
Yuda
K
Suh
Y-A
Kumar
PKR
Nishikawa
F
Maeda
H
Taira
K
Ohtsuka
E
Nishikawa
S
Constructing an efficient trans-acting genomic HDV ribozyme.
FEBS Lett
394
1996
132
68
Wagner
RW
Gene inhibition using antisense oligodeoxynucleotides.
Nature
372
1994
333
69
Lodish H, Baltimore D, Berk A, Zipursky SL, Matsudaira P, Darnell J: Molecular Cell Biology. New York, NY, Scientific American, 1995, p 641
70
Kobayashi
H
Dorai
T
Holland
JF
Ohnuma
T
Reversal of drug sensitivity in multidrug-resistant tumor cells by an MDR1 (PGY1) ribozyme.
Cancer Res
54
1994
1271
71
Kiehntopf
M
Brach
MA
Licht
T
Petschauer
S
Karawajew
L
Kirschning
C
Herrmann
F
Ribozyme-mediated cleavage of the MDR-1 transcript restores chemosensitivity in previously resistant cancer cells.
EMBO J
13
1994
4645
72
Kashani-Sabet
M
Funato
T
Florenes
VA
Fodstad
O
Scanlon
KJ
Suppression of the neoplastic phenotype in vivo by an anti-ras ribozyme.
Cancer Res
54
1994
900
73
Ohta
Y
Kijima
H
Ohkawa
T
Kashani-Sabet
M
Scanlon
KJ
Tissue-specific expression of an anti-ras ribozyme inhibits proliferation of human malignant melanoma cells.
Nucleic Acids Res
24
1996
938
74
Lange
W
Cantin
EM
Finke
J
Dolken
G
In vitro and in vivo effects of synthetic ribozymes targeted against BCR/ABL mRNA.
Leukemia
7
1993
1786
75
Snyder
DS
Wu
Y
Wang
JL
Rossi
JJ
Swiderski
P
Kaplan
BE
Forman
SJ
Ribozyme-mediated inhibition of bcr-abl gene expression in a Philadelphia chromosome-positive cell line.
Blood
82
1993
600
76
Leopold
LH
Shore
SK
Newkirk
TA
Reddy
RMV
Reddy
EP
Multi-unit ribozyme-mediated cleavage of bcr-abl mRNA in myeloid leukemias.
Blood
85
1995
2162
77
(abstr, suppl 1)
Mills
KI
Walsh
V
Gilkes
AF
Goringe
A
Twomey
C
James
HA
Gibson
I
In vitro ribozyme treatment of 32D cells expressing a BCR-ABL construct prolongs the survival of SCID mice.
Blood
88
1996
577a
78
Czubayko
F
Schulte
AM
Berchem
GJ
Wellstein
A
Melanoma angiogenesis and metastasis modulated by ribozyme targeting of the secreted growth factor pleiotrophin.
Proc Natl Acad Sci USA
93
1996
14753
79
Zhou
C
Bahner
IC
Larson
GP
Zaia
JA
Rossi
JJ
Kohn
DB
Inhibition of HIV-1 in human T-lymphocytes by retrovirally transduced anti-tat and rev hammerhead ribozymes.
Gene
149
1994
33
80
Leavitt
MC
Yu
M
Wong-Staal
F
Looney
DJ
Ex vivo transduction and expansion of CD4+ lymphocytes from HIV+ donors: Prelude to a ribozyme gene therapy trial.
Gene Ther
3
1996
599
81
Bauer
G
Valdez
P
Kearns
K
Bahner
I
Wen
SF
Zaia
JA
Kohn
DB
Inhibition of human immunodeficiency virus-1 (HIV-1) replication after transduction of granulocyte colony-stimulating factor-mobilized CD34(+) cells from HIV-1–infected donors using retroviral vectors containing anti–HIV-1 genes.
Blood
89
1997
2259
82
Beck
J
Nassal
M
Efficient hammerhead ribozyme-mediated cleavage of the structured hepatitis B virus encapsidation signal in vitro and in cell extracts, but not in intact cells.
Nucleic Acids Res
23
1995
4954
83
Welch
PJ
Tritz
R
Yei
S
Leavitt
M
Yu
M
Barber
J
A potential therapeutic application of hairpin ribozymes: In vitro and in vivo studies of gene therapy for hepatitis C virus infection.
Gene Ther
3
1996
994
84
Flory
CM
Pavco
PA
Jarvis
TC
Lesch
ME
Wincott
FE
Beigelman
L
Hunt
SW
III
Schrier
DJ
Nuclease-resistant ribozymes decrease stromelysin mRNA levels in rabbit synovium following exogenous delivery to the knee joint.
Proc Natl Acad Sci USA
93
1996
754
85
Cameron
FH
Jennings
PA
Multiple domains in a ribozyme construct confer increased suppressive activity in monkey cells.
Antisense Res Dev
4
1994
87
86
Steinecke
P
Herget
T
Schreier
PH
Expression of a chimeric ribozyme gene results in endonucleolytic cleavage of target mRNA and a concomitant reduction of gene expression in vivo.
EMBO J
11
1992
1525
87
Davies
C
Haseloff
J
Symons
RH
Structure, self-cleavage and replication of two viroid-like satellite RNAs (virusoids) of subterranean clover mottle virus.
Virology
177
1990
216
88
Heidenreich
O
Xu
X
Nerenberg
M
A hammerhead ribozyme cleaves its target RNA during RNA preparation.
Antisense Nucleic Acid Drug Dev
6
1996
141
89
Ohta
Y
Kijima
H
Kashani-Sabet
M
Scanlon
KJ
Suppression of the malignant phenotype of melanoma cells by anti-oncogene ribozymes.
J Invest Dermatol
106
1996
275
90
James
H
Mills
K
Gibson
I
Investigating and improving the specificity of ribozymes directed against the bcr-abl translocation.
Leukemia
10
1996
1054
91
Lange
W
Daskalakis
M
Finke
J
Dolken
G
Comparison of different ribozymes for efficient and specific cleavage of BCR/ABL related mRNAs.
FEBS Lett
338
1994
175
92
Wright
L
Wilson
SB
Milliken
S
Biggs
J
Kearney
P
Ribozyme-mediated cleavage of the bcr/abl transcript expressed in chronic myeloid leukemia.
Exp Hematol
21
1993
1714
93
Shore
SK
Nabissa
PM
Reddy
EP
Ribozyme-mediated cleavage of the BCRABL oncogene transcript: In vitro cleavage of RNA and in vivo loss of p210 protein-kinase activity.
Oncogene
8
1993
3183
94
Kozu
T
Sueoka
E
Okabe
S
Sueoka
N
Komori
A
Fujiki
H
Designing of chimeric DNA/RNA hammerhead ribozymes to be targeted against AML1/MTG8 mRNA.
J Cancer Res Clin Oncol
122
1996
254
95
Weerasinghe
M
Liem
SE
Asad
S
Read
SE
Joshi
S
Resistance to human immunodeficiency virus type 1 (HIV-1) infection in human CD4+ lymphocyte-derived cell lines conferred by using retroviral vectors expressing an HIV-1 RNA-specific ribozyme.
J Virol
65
1991
5531
96
Lo
KMS
Biasolo
MA
Dehni
G
Palu
G
Haseltine
WA
Inhibition of replication of HIV-1 by retroviral vectors expressing tat-antisense and anti-tat ribozyme RNA.
Virology
190
1992
176
97
Yamada
O
Yu
M
Yee
J-K
Kraus
G
Looney
D
Wong-Staal
F
Intracellular immunization of human T cells with a hairpin ribozyme against human immunodeficiency virus type I.
Gene Ther
1
1994
38
98
Zhou
C
Bahner
I
Rossi
JJ
Kohn
DB
Expression of hammerhead ribozymes by retroviral vectors to inhibit HIV-1 replication: Comparison of RNA levels and viral inhibition.
Antisense Nucleic Acid Drug Dev
6
1996
17
99
Wong-Staal F, Poeschla E, Looney D, Yu M, Leavitt M, Yamada O, Yee J-K: RAC Submission # 9309-057. 1993
100
Lieber
A
He
CY
Polyak
SJ
Gretch
DR
Barr
D
Kay
MA
Elimination of hepatitis C virus RNA in infected human hepatocytes by adenovirus-mediated expression of ribozymes.
J Virol
70
1996
8782
101
Sakamoto
N
Wu
CH
Wu
GY
Intracellular cleavage of hepatitis C virus RNA and inhibition of viral protein translation by hammerhead ribozymes.
J Clin Invest
98
1996
2720
102
Tang
XB
Hobom
G
Luo
D
Ribozyme mediated destruction of influenza A virus in vitro and in vivo.
J Med Virol
42
1994
385
103
Larsson
S
Hotchkiss
G
Andang
M
Nyholm
T
Inzunza
J
Jansson
I
Ahrlund-Richter
L
Reduced β2 microglobulin mRNA levels in transgenic mice expressing a designed hammerhead ribozyme.
Nucleic Acids Res
22
1994
2242
104
Lieber
A
Kay
MA
Adenovirus-mediated expression of ribozymes in mice.
J Virol
70
1996
3153
105
Desjardins
JP
Sproat
BS
Beijer
B
Blaschke
M
Dunkel
M
Gerdes
W
Ludwig
J
Reither
V
Rupp
T
Iversen
PL
Pharmacokinetics of a synthetic, chemically modified hammerhead ribozyme against the rat cytochrome P-450 3A2 mRNA after single intravenous injections.
J Pharmacol Exp Ther
278
1996
1419
106
Lyngstadaas
SP
Risnes
S
Sproat
BS
Thrane
PS
Prydz
HP
A synthetic, chemically modified ribozyme eliminates amelogenin, the major translation product in developing mouse enamel in vivo.
EMBO J
14
1995
5224
107
Sokol
DL
Murray
JD
Antisense and ribozyme constructs in transgenic animals.
Transgenic Res
5
1996
363
108
Zhao
JJ
Pick
L
Generating loss-of-function phenotypes of the fushi tarazu gene with a targeted ribozyme in Drosophila.
Nature
365
1993
448
109
Scanlon
KJ
Jiao
L
Funato
T
Wang
W
Tone
T
Rossi
JJ
Kashani-Sabet
M
Ribozyme-mediated cleavage of c-fos mRNA reduces gene expression of DNA synthesis enzymes and metallothionein.
Proc Natl Acad Sci USA
88
1991
10591
110
Turck
J
Pollock
AS
Lee
LK
Marti
H-P
Lovett
DH
Matrix metalloproteinase 2 (gelatinase A) regulates glomerular mesangial cell proliferation and differentiation.
J Biol Chem
271
1996
15074
111
Mazzolini
L
Axelos
M
Lescure
N
Yot
P
Assaying synthetic ribozymes in plants: high-level expression of a functional hammerhead structure fails to inhibit target gene activity in transiently transformed protoplasts.
Plant Mol Biol
20
1992
715
112
Perriman
R
Graf
L
Gerlach
WL
A ribozyme that enhances gene suppression in tobacco protoplasts.
Antisense Res Dev
3
1993
253
113
Atkins
D
Gerlach
WL
Artificial ribozyme and antisense gene expression in Saccharomyces cerevisiae.
Antisense Res Dev
4
1994
109
114
Atkins
D
Patrikakis
M
Izant
JG
The ade6 gene of the fission yeast as a target for antisense and ribozyme RNA-mediated suppression.
Antisense Res Dev
5
1995
295
115
McIntyre
CL
Bettenay
HM
Manners
JM
Strategies for the suppression of peroxidase gene expression in tobacco. II. In vivo suppression of peroxidase activity in transgenic tobacco using ribozyme and antisense constructs.
Transgenic Res
5
1996
263
116
Turner PC (Ed): Methods in Molecular Biology 74: Ribozyme Protocols. Totowa, NJ, Humana, 1997
117
Gibson I (Ed): Antisense and Ribozyme Methodology. Laboratory Companion. Weinheim, Germany, Chapman & Hall GmbH, 1997
118
Zoumadakis
M
Neubert
WJ
Tabler
M
The influence of imperfectly paired helices I and III on the catalytic activity of hammerhead ribozymes.
Nucleic Acids Res
22
1994
5271
119
Xing
Z
Whitton
JL
Ribozymes which cleave Arenavirus RNAs: Identification of susceptible target sites and inhibition by target site secondary structure.
J Virol
66
1992
1361
120
Kronenwett
R
Haas
R
Sczakiel
G
Kinetic selectivity of complementary nucleic acids: bcr-abl-directed antisense RNA and ribozymes.
J Mol Biol
259
1996
632
121
Auron
PE
Rindone
WP
Vary
CPH
Celentano
JJ
Vournakis
JN
Computer-aided prediction of RNA secondary structures.
Nucleic Acids Res
10
1982
403
122
Jacobson
AB
Good
L
Simonetti
J
Zuker
M
Some simple computational methods to improve the folding of large RNAs.
Nucleic Acids Res
12
1984
45
123
Herschlag
D
Implications of ribozyme kinetics for targeting the cleavage of specific RNA molecules in vivo: More isn't always better.
Proc Natl Acad Sci USA
88
1991
6921
124
Sioud
M
Effects of variations in length of hammerhead ribozyme antisense arms upon the cleavage of longer RNA substrates.
Nucleic Acids Res
25
1997
333
125
Tabler
M
Homann
M
Tzortzakaki
S
Sczakiel
G
A three-nucleotide helix I is sufficient for full activity of a hammerhead ribozyme: Advantages of an asymmetric design.
Nucleic Acids Res
22
1994
3958
126
Crisell
P
Thompson
S
James
W
Inhibition of HIV-1 replication by ribozymes that show poor activity in vitro.
Nucleic Acids Res
21
1993
5251
127
Hormes
R
Homann
M
Oelze
I
Marschall
P
Tabler
M
Eckstein
F
Sczakiel
G
The subcellular localization and length of hammerhead ribozymes determine efficacy in human cells.
Nucleic Acids Res
25
1997
769
128
Chapman
KB
Szostak
JW
In vitro selection of catalytic RNAs.
Curr Opin Sruct Biol
4
1994
618
129
Joyce
GF
In vitro evolution of nucleic acids.
Curr Opin Sruct Biol
4
1994
331
130
Bergan
R
Connell
Y
Fahmy
B
Kyle
E
Neckers
L
Aptameric inhibition of p210bcr-abl tyrosine kinase autophosphorylation by oligodeoxynucleotides of defined sequence and backbone structure.
Nucleic Acids Res
22
1994
2150
131
Bergan
RC
Kyle
E
Connell
Y
Neckers
L
Inhibition of protein-tyrosine kinase activity in intact cells by the aptameric action of oligodeoxynucleotides.
Antisense Res Dev
5
1995
33
132
Stull
RA
Szoka
FC
Jr
Antigene, ribozyme and aptamer nucleic acid drugs: Progress and prospects.
Pharm Res
12
1995
465
133
Chen
C-J
Banerjea
AC
Harmison
GG
Haglund
K
Schubert
M
Multitarget-ribozyme directed to cleave at up to nine highly conserved HIV-1 env RNA regions inhibits HIV-1 replication—Potential effectiveness against most presently sequenced HIV-1 isolates.
Nucleic Acids Res
20
1992
4581
134
Leopold
LH
Shore
SK
Reddy
EP
Multi-unit anti-BCR-ABL ribozyme therapy in chronic myelogenous leukemia.
Leuk Lymphoma
22
1996
365
135
Pieken
WA
Olsen
DB
Benseler
F
Aurup
H
Eckstein
F
Kinetic characterization of ribonuclease-resistant 2′-modified hammerhead ribozymes.
Science
253
1991
314
136
Paolella
G
Sproat
BS
Lamond
AI
Nuclease resistant ribozymes with high catalytic activity.
EMBO J
11
1992
1913
137
Bertrand
E
Castanotto
D
Zhou
C
Carbonnelle
C
Lee
NS
Good
P
Chatterjee
S
Grange
T
Pictet
R
Kohn
D
Engelke
D
Rossi
JJ
The expression cassette determines the functional acivity of ribozymes in mammalian cells by controlling their intracellular localization.
RNA
3
1997
75
138
Sullenger
BA
Ribozyme-mediated repair of RNAs encoding mutant tumor suppressors.
Cytokines Mol Ther
2
1996
201
139
Sarver
N
Cantin
EM
Chang
PS
Zaia
JA
Ladne
PA
Stephens
DA
Rossi
JJ
Ribozymes as potential anti-HIV-1 therapeutic agents.
Science
247
1990
1222
140
Good
PD
Krikos
AJ
Li
SXL
Bertrand
E
Lee
NS
Giver
L
Ellington
A
Zaia
JA
Rossi
JJ
Engelke
DR
Expression of small, therapeutic RNAs in human cell nuclei.
Gene Ther
4
1997
45
141
Thompson
JD
Ayers
DF
Malmstrom
TA
McKenzie
TL
Ganousis
L
Chowrira
BM
Couture
L
Stinchcomb
DT
Improved accumulation and activity of ribozymes expressed from a tRNA-based RNA polymerase III promoter.
Nucleic Acids Res
23
1995
2259
142
Sullenger
BA
Cech
TR
Tethering ribozymes to a retroviral packaging signal for destruction of viral RNA.
Science
262
1993
1566
143
Lawrence
JB
Singer
RH
Intracellular localization of messenger RNAs for cytoskeletal proteins.
Cell
45
1986
407
144
Ding
D
Lipshitz
HD
Localized RNAs and their functions.
Bioessays
15
1993
651
145
Twomey CM, Gibson I: The intracellular distribution of fluorescently labeled ribozyme in the 32Db3a2 murine cell line, in Molecular and Cellular Biology of Gene Therapy. Keystone Symposia. Snowbird, UT, 1997, p 71 (abstr)
146
Savelyeva NN, Gibson I: Unprotected ribozymes against c-myc mRNA cleave the RNA in nuclei isolated from HL-60 cells, in Molecular and Cellular Biology of Gene Therapy. Keystone Symposia. Snowbird, UT, 1997, p 68 (abstr)
147
Bertrand
E
Fromont-Racine
M
Pictet
R
Grange
T
Visualisation of the interaction of a regulatory protein with RNA in vivo.
Proc Natl Acad Sci USA
90
1993
3496
148
(editorial)
Stein
CA
Krieg
A
Problems in the interpretation of data derived from the in vitro and in vivo use of antisense oligodeoxynucleotides.
Antisense Res Dev
4
1994
67
149
Yaswen
P
Stampfer
MR
Ghosh
K
Cohen
JS
Effects of sequence of thioated oligonucleotides on cultured human mammary epithelial cells.
Antisense Res Dev
3
1993
67
150
Vaerman
JL
Lammineur
C
Moureau
P
Lewalle
P
Deldime
F
Blumenfeld
M
Martiat
P
BCR-ABL antisense oligodeoxyribonucleotides suppress the growth of leukemic and normal hematopoietic cells by a sequence-specific but nonantisense mechanism.
Blood
86
1995
3891
151
von Ahsen
U
Davies
J
Schroeder
R
Antibiotic inhibition of group I ribozyme function.
Nature
353
1991
368
152
Stage
TK
Hertel
KJ
Uhlenbeck
OC
Inhibition of the hammerhead ribozyme by neomycin.
RNA
1
1995
95
153
Murray
JB
Arnold
JRP
Antibiotic interactions with the hammerhead ribozyme—Tetracyclines as a new class of hammerhead inhibitor.
Biochem J
317
1996
855
154
Rogers
J
Chang
AH
von Ahsen
U
Schroeder
R
Davies
J
Inhibition of the self-cleavage reaction of the human hepatitis delta virus ribozyme by antibiotics.
J Mol Biol
259
1996
916
155
Olive
JE
De Abreu
DM
Rastogi
T
Andersen
AA
Mittermaier
AK
Beattie
TL
Collins
RA
Enhancement of Neurospora VS ribozyme cleavage by tuberactinomycin antibiotics.
EMBO J
14
1995
3247
Sign in via your Institution