throbber
Physiol. Res. 50: 536-546, 2001
`
`MINIREVIEW
`
`The Mechanism of Alloxan and Streptozotocin Action in
`B Cells of the Rat Pancreas
`T. SZKUDELSKI
`
`Department of Animal Physiology and Biochemistry, University of Agriculture, Poznan, Poland
`
`Received November 2, 2000
`Accepted March 20, 2001
`
`Summary
`Alloxan and streptozotocin are widely used to induce experimental diabetes in animals. The mechanism of their action
`in B cells of the pancreas has been intensively investigated and now is quite well understood. The cytotoxic action of
`both these diabetogenic agents is mediated by reactive oxygen species, however, the source of their generation is
`different in the case of alloxan and streptozotocin. Alloxan and the product of its reduction, dialuric acid, establish a
`redox cycle with the formation of superoxide radicals. These radicals undergo dismutation to hydrogen peroxide.
`Thereafter highly reactive hydroxyl radicals are formed by the Fenton reaction. The action of reactive oxygen species
`with a simultaneous massive increase in cytosolic calcium concentration causes rapid destruction of B cells.
`Streptozotocin enters the B cell via a glucose transporter (GLUT2) and causes alkylation of DNA. DNA damage
`induces activation of poly ADP-ribosylation, a process that is more important for the diabetogenicity of streptozotocin
`than DNA damage itself. Poly ADP-ribosylation leads to depletion of cellular NAD+ and ATP. Enhanced ATP
`dephosphorylation after streptozotocin treatment supplies a substrate for xanthine oxidase resulting in the formation of
`superoxide radicals. Consequently, hydrogen peroxide and hydroxyl radicals are also generated. Furthermore,
`streptozotocin liberates toxic amounts of nitric oxide that inhibits aconitase activity and participates in DNA damage.
`As a result of the streptozotocin action, B cells undergo the destruction by necrosis.
`
`Key words
`Alloxan • Streptozotocin • Pancreatic B cells • Mechanism of action • Diabetes
`
`The induction of experimental diabetes in the rat
`using chemicals which selectively destroy pancreatic
`B cells is very convenient and simple to use. The most
`usual substances to induce diabetes in the rat are alloxan
`and streptozotocin. The understanding of changes in
`
`B cells of the pancreas as well as in the whole organism
`after alloxan or streptozotocin treatment is essential for
`using these compounds as diabetogenic agents. The
`metabolic disturbances in alloxan- and streptozotocin-
`treated rats were described recently by Szkudelski et al.
`
` ISSN 0862-8408
`PHYSIOLOGICAL RESEARCH
` 2001 Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
` Fax+420 224920590
`E-mail physres@biomed.cas.cz
` http://www.biomed.cas.cz/physiolres/s.htm
`
`NOVARTIS EXHIBIT 2090
`Par v. Novartis, IPR 2016-01479
`Page 1 of 10
`
`

`

`538
`
` Szkudelski
`
`Vol. 50
`
`(1998). This review focuses on the elucidation of the
`mechanism of cytotoxic action of alloxan and
`streptozotocin in B cells of the rat pancreas.
`
`1. The mechanism of alloxan action
`
`Alloxan (2,4,5,6-tetraoxypyrimidine; 5,6-dioxyura-
`cil) was first described by Brugnatelli in 1818. Wöhler
`and Liebig used the name “alloxan” and described its
`synthesis by uric acid oxidation (for review see Lenzen
`and Panten 1988). The diabetogenic properties of this
`drug were reported many years later by Dunn, Sheehan
`and McLethie (1943), who studied the effect of its
`administration in rabbits and reported a specific necrosis
`of pancreatic islets. Since then, alloxan diabetes has been
`commonly utilized as an animal model of insulin-
`dependent diabetes mellitus (IDDM).
`Alloxan exerts its diabetogenic action when it is
`administered parenterally:
`intravenously,
`intraperito-
`neally or subcutaneously. The dose of alloxan required
`for inducing diabetes depends on the animal species,
`route of administration and nutritional status. Human
`islets are considerably more resistant to alloxan than
`those of the rat and mouse (Eizirik et al. 1994). The most
`frequently used intravenous dose of this drug to induce
`diabetes in rats is 65 mg/kg b.w. (Gruppuso et al. 1990,
`Boylan et al. 1992). When alloxan is given intraperito-
`nealy or subcutaneously its effective dose must be 2-3
`times higher. The intraperitoneal dose below 150 mg/kg
`b.w. may be insufficient for inducing diabetes in the rat
`(Katsumata et al. 1992, 1993). Fasted animals are more
`susceptible to alloxan (Katsumata et al. 1992, Szkudelski
`et al. 1998), whereas increased blood glucose provides
`partial protection (Bansal et al. 1980, Szkudelski et al.
`1998).
`The mechanism of alloxan action has been
`intensively studied, predominantly in vitro, and is now
`characterized quite well. Using isolated islets (Weaver et
`al. 1978b) and perfused rat pancreas (Kliber et al. 1996)
`it was demonstrated that alloxan evokes a sudden rise in
`insulin secretion in the presence or absence of glucose.
`This phenomenon appeared just after alloxan treatment
`and was not observed after repetitive exposure of islets to
`this diabetogenic agent (Weaver et al. 1978b). The
`sudden rise in blood insulin concentration was also
`observed in vivo just after alloxan injection to rats
`(Szkudelski et al. 1998). Alloxan-induced insulin release
`is, however, of short duration and is followed by
`complete suppression of the islet response to glucose,
`
`even when high concentrations (16.6 mM) of this sugar
`were used (Kliber et al. 1996).
`Alloxan is a hydrophilic and unstable substance. Its
`half-life at neutral pH and 37 °C is about 1.5 min and is
`longer at lower temperatures (Lenzen and Munday 1991).
`On the other hand, when a diabetogenic dose is used, the
`time of alloxan decomposition is sufficient to allow it to
`reach the pancreas in amounts that are deleterious.
`The action of alloxan in the pancreas is preceded by
`its rapid uptake by the B cells (Weaver et al. 1978a,
`Boquist et al. 1983). Rapid uptake by insulin-secreting
`cells has been proposed to be one of the important
`features determining alloxan diabetogenicity. Another
`aspect concerns the formation of reactive oxygen species
`(Heikkila et al. 1976). A similar uptake of alloxan also
`takes place in the liver. However, the liver and other
`tissues are more resistant to reactive oxygen species in
`comparison to pancreatic B cells and this resistance
`protects them against alloxan toxicity (Malaisse et al.
`1982, Tiedge et al. 1997). The formation of reactive
`oxygen species is preceded by alloxan reduction. In
`B cells of the pancreas its reduction occurs in the
`presence of different reducing agents. Since alloxan
`exhibits a high affinity to the SH-containing cellular
`compounds, reduced glutathione (GSH), cysteine and
`protein-bound
`sulfhydryl
`groups
`(including SH-
`containing enzymes) are very susceptible to its action
`(Lenzen and Munday 1991). However, other reducing
`agents such as ascorbate may also participate in this
`reduction (Zhang et al. 1992). Lenzen et al. (1987)
`proposed that one of the SH-containing compounds
`essential for proper glucose-induced insulin secretion is
`glucokinase (EC 2.7.1.2), being very vulnerable to
`alloxan. Alloxan reacts with two -SH groups in the sugar-
`binding side of glucokinase resulting in the formation of
`the disulfide bond and inactivation of the enzyme.
`Glucose can protect glucokinase against the inactivation
`hindering the access of alloxan to the -SH groups of the
`enzyme (Lenzen et al. 1987, 1988, Lenzen and Mirzaie-
`Petri 1991).
`Dialuric acid is formed as a result of alloxan
`reduction. It
`is
`then re-oxidized back
`to alloxan
`establishing a redox cycle for
`the generation of
`superoxide
`radicals
`(Munday 1988). The
`reaction
`between alloxan and dialuric acid is a process in which
`intermediate alloxan radicals (HA•) and an unidentified
`"compound 305" (maximum absorption at 305 nm) is
`formed. The latter appears when alloxan is reduced by
`GSH (Sakurai and Ogiso 1991). Superoxide radicals are
`
`NOVARTIS EXHIBIT 2090
`Par v. Novartis, IPR 2016-01479
`Page 2 of 10
`
`

`

`2001
`
`Alloxan and Streptozotocin Action
`
`539
`
`able to liberate ferric ions from ferritin and reduce them
`to ferrous ions. Fe3+ can also be reduced by alloxan
`radicals (Sakurai and Ogiso 1995). Moreover, superoxide
`radicals undergo dismutation to hydrogen peroxide:
`•− + O2
`•− + 2 H+ → H2O2 + O2
`O2
`This reaction may occur spontaneously or may be
`catalyzed by superoxide dismutase
`(EC 1.15.1.1)
`
`(Malaisse 1982). In the presence of Fe2+ and hydrogen
`peroxide, highly reactive hydroxyl radicals are then
`formed according to the Fenton reaction (Fig. 1):
`Fe2+ + H2O2 → Fe3+ + OH− + OH•−
`The action of hydroxyl radicals following alloxan
`treatment was demonstrated in vitro (Grankvist 1981,
`Munday 1988) and in vivo (Kurahashi et al. 1993).
`
`Fig. 1. The mechanism of alloxan-induced
`reactive oxygen species generation in B cells of
`rat pancreas. GKa, GKi – glucokinase active
`and inactive, respectively; HA• – alloxan
`radicals; [Ca2+]i –
`intracellular calcium
`concentration.
`
`One of the targets of the reactive oxygen species
`is DNA of pancreatic islets. Its fragmentation takes place
`in B cells exposed to alloxan (Takasu et al. 1991a,
`Sakurai and Ogiso 1995). DNA damage stimulates poly
`ADP-ribosylation, a process participating in DNA repair.
`Some inhibitors of poly ADP-ribosylation can partially
`restrict alloxan
`toxicity. This effect
`is, however,
`suggested to be due to their ability to scavenge free
`radicals rather than to a restriction of poly ADP-
`ribosylation initiated by alloxan (Sandler and Swenne
`1983, LeDoux et al. 1988). Superoxide dismutase,
`catalase (EC 1.11.1.6) (Grankvist et al. 1979, Grankvist
`1981, Jörns et al. 1999) and non-enzymatic scavengers of
`hydroxyl radicals (Ebelt et al. 2000) were also found to
`protect against alloxan toxicity. Therefore, chemicals
`rendering anti-oxidative properties and inhibiting poly
`ADP-ribosylation can attenuate alloxan toxicity.
`It has been argued that glucose counteracts
`alloxan cytotoxicity in vitro and in vivo. This ability,
`however, is not only the result of the protection of
`glucokinase. The protective effect of glucose against
`necrotic death of B cells may be due to interaction of the
`sugar with the glucose transporter GLUT2 resulting in
`limited alloxan uptake (Jörns et al. 1997).
`
` It has been previously proposed that the action
`of glucose is also related to its metabolism and to the
`increased generation of reducing equivalents (NADH and
`NADPH) accelerating the recirculation of glutathione.
`GSH is known to provide protection against free radicals
`(Donnini et al. 1996). It may thus divert hydrogen
`peroxide from the pathway leading to the formation of
`hydroxyl radicals (Malaisse 1982, Malaisse-Lagae et al.
`1983, Pipeleers and van de Winkel 1986):
`GSSG + 2 NADPH → 2 GSH + 2 NADP+
`H2O2 + 2 GSH → GSSG + 2 H2O
`Moreover, Sakurai and Ogiso (1991) observed
`that the in vitro generation of hydroxyl radicals in the
`presence of alloxan
`strongly depends on GSH
`concentration. GSH in low concentrations potentiated the
`formation of
`these
`radicals, whereas
`the oxygen
`consumption, autoxidation of dialuric acid and formation
`of hydroxyl radicals were significantly inhibited in higher
`concentrations. GSH at high concentrations can also
`inhibit HA• generation and directly neutralize hydroxyl
`radicals. Thiyl radicals (GS•) formed in this reaction are
`then converted to GSSG:
`GSH + OH•− → GS• + H2O
`GS• + GS• → GSSG
`
`NOVARTIS EXHIBIT 2090
`Par v. Novartis, IPR 2016-01479
`Page 3 of 10
`
`

`

`540
`
` Szkudelski
`
`Vol. 50
`
`Indeed, in rat islets incubated with alloxan the
`GSH content and GSH/GSSG ratio were decreased
`(Malaisse et al. 1982), whereas glucose evoked the
`opposite effect.
`In the in vivo experiment, glucose given to rats
`20 min prior to alloxan partially restricted alloxan-
`induced increase in the activity of glutathione peroxidase
`(EC 1.11.1.9) and mitigated the drop of liver nonprotein
`-SH groups (especially reduced glutathione) (Szkudelski
`et al. 1998). The protective action of this sugar is,
`however, strongly glucose and alloxan dose-dependent
`(Harman and Fischer 1982, Gorray et al. 1983).
`in
`It has been proposed
`that disturbances
`intracellular calcium homeostasis constitute an important
`step in the diabetogenic action of alloxan. This concept
`was confirmed by in vitro and in vivo experiments
`demonstrating that alloxan elevates cytosolic free Ca2+
`concentration in pancreatic B cells (Kim et al. 1994, Park
`et al. 1995). This effect arises from several events:
`alloxan-induced calcium influx from extracellular fluid,
`exaggerated calcium mobilization from
`intracellular
`stores and its limited elimination from the cytoplasm. The
`calcium influx may result from the ability of alloxan to
`depolarize pancreatic B cells (Dean and Matthews 1972).
`Depolarization of the cell membrane opens voltage-
`dependent calcium channels and enhances calcium entry
`into cells. Alloxan was also found to exert a stimulatory
`effect on mitochondrial Ca2+ efflux with simultaneous
`inhibitory action on Ca2+ uptake by mitochondria (Nelson
`and Boquist 1982, Lenzen et al. 1992). The restriction of
`calcium removal from the cells due to alloxan-induced
`inhibition of liver plasma membrane Ca2+-ATPase was
`also reported (Seckin et al. 1993). The effect of alloxan
`on intracellular calcium concentration seems to be
`mediated, at least partially, by H2O2 since hydrogen
`peroxide
`itself exerts a similar effect on calcium
`concentration in B cells (Park et al. 1995).
`Thus, the previously mentioned sudden rise in
`insulin release from B cells treated with alloxan (Weaver
`et al. 1978b, Kliber et al. 1996) may be one of the effects
`in cytosolic Ca2+
`of alloxan-induced augmentation
`concentration (Weaver et al. 1978b, Kim et al. 1994).
`The exaggerated concentration of this ion contributes to
`supraphysiological insulin release and, together with
`reactive oxygen species, causes damage of pancreatic
`B cells.
`
`The results of experiments with calcium channel
`antagonists have confirmed
`the
`important role of
`cytosolic calcium in the cytotoxic action of alloxan.
`
`the
`Pretreatment of rats with verapamil prevented
`alloxan-induced increase in B cell Ca2+ concentration and
`abolished the stimulatory effect of alloxan on insulin
`release (Kim et al. 1994). The calcium channel
`antagonists (verapamil and diltiazem) also suppressed
`hyperglycemia and the onset of alloxan diabetes in rats
`(Katsumata et al. 1992, Kim et al. 1994).
`Summing up, the toxic action of alloxan on
`pancreatic B cells, described many years ago by Dunn et
`al. (1943), are the sum of several processes such as
`oxidation of essential
`-SH groups,
`inhibition of
`glucokinase, generation of free radicals and disturbances
`in intracellular calcium homeostasis.
`Many investigators suggested that the selectivity
`of alloxan action is not quite satisfactory. Recent
`experiments confirmed this objection. The diabetogenic
`dose of alloxan was found to decrease -SH groups
`accompanied by a simultaneous rise in glutathione
`peroxidase activity in the rat liver two minutes after its
`administration (Szkudelski et al. 1998). At the same time,
`the blood insulin concentration rose dramatically. This
`exaggerated insulinemia did not evoke, however, any
`significant
`reduction of blood glucose suggesting
`impaired peripheral insulin sensitivity in the short time
`after alloxan treatment (Szkudelski et al. 1998). It was
`also observed
`that alloxan
`intensified basal and
`epinephrine-induced lipolysis in isolated rat adipocytes
`and insulin failed to restrict this effect (Kandulska et al.
`1999).
`
`Thus, using alloxan to evoke diabetes, animals
`should be examined after proper period of time to
`minimize side effects of alloxan action. It should also be
`emphasized that the range of the diabetogenic dose of
`alloxan is quite narrow and even light overdosing may be
`generally toxic causing the loss of many animals. This
`loss is most likely due to kidney tubular cell necrotic
`toxicity, in particular when too high doses of alloxan are
`administered (Lenzen et al. 1996).
`
`2. The mechanism of streptozotocin action
`
`2-deoxy-2-(3-(methyl-3-
`(STZ,
`Streptozotocin
`nitrosoureido)-D-glucopyranose)
`is
`synthesized by
`Streptomycetes achromogenes and is used to induce both
`insulin-dependent and non-insulin-dependent diabetes
`mellitus (IDDM and NIDDM, respectively).
`The range of the STZ dose is not as narrow as in the
`case of alloxan. The frequently used single intravenous
`dose in adult rats to induce IDDM is between 40 and 60
`
`NOVARTIS EXHIBIT 2090
`Par v. Novartis, IPR 2016-01479
`Page 4 of 10
`
`

`

`2001
`
`Alloxan and Streptozotocin Action
`
`541
`
`mg/kg b.w. (Ganda et al. 1976), but higher doses are also
`used. STZ
`is also efficacious after
`intraperitoneal
`administration of a similar or higher dose, but single dose
`below 40 mg/kg b.w. may be ineffective (Katsumata et
`al. 1992). For instance, when 50 mg/kg b.w. STZ are
`injected
`intravenously
`to
`fed
`rats, blood glucose
`(determined 2 weeks after treatment) can reach about 15
`mM (Szkudelski, unpublished observations).
`STZ may also be given in multiple low doses.
`Such treatment is used predominantly in the mouse and
`the induction of IDDM is mediated by the activation of
`immune mechanisms. However, Ziegler et al. (1984) and
`Wright and Lacy (1988) demonstrated that the non-
`specific activation of the immune system via complete
`Freund's adjuvant prior to STZ injections allows to
`reduce its diabetogenic dose even in the rat.
`NIDDM can easily be induced in rats by
`intravenous or intraperitoneal treatment with 100 mg/kg
`b.w. STZ on the day of birth. This method of NIDDM
`induction was described for the first time by Portha et al.
`(1974). At 8-10 weeks of age and thereafter, rats
`neonatally
`treated with STZ manifest mild basal
`hyperglycemia, an impaired response to the glucose
`tolerance test (Portha et al. 1979) and a loss of B cell
`sensitivity to glucose (Giroix et al. 1983).
`Streptozotocin action in B cells is accompanied
`by characteristic alterations in blood insulin and glucose
`concentrations. Two
`hours
`after
`injection,
`the
`hyperglycemia is observed with a concomitant drop in
`blood insulin. About six hours later, hypoglycemia occurs
`with high levels of blood insulin. Finally, hyperglycemia
`develops and blood insulin levels decrease (West et al.
`1996). These changes in blood glucose and insulin
`concentrations reflect abnormalities in B cell function.
`STZ impairs glucose oxidation (Bedoya et al. 1996) and
`decreases insulin biosynthesis and secretion (Bolaffi et al.
`1987, Nukatsuka et al. 1990b). It was observed that STZ
`at first abolished
`the B cell response
`to glucose.
`Temporary return of responsiveness then appears which
`is followed by its permanent loss and cells are damaged
`(West et al. 1996).
`STZ is taken up by pancreatic B cells via
`glucose transporter GLUT2. A reduced expression of
`GLUT2 has been found to prevent the diabetogenic
`action of STZ (Schnedl et al. 1994, Thulesen et al. 1997).
`Wang and Gleichmann (1995, 1998) observed that STZ
`itself restricts GLUT2 expression in vivo and in vitro
`when administered in multiple doses.
`Intracellular action of STZ results in changes of
`DNA in pancreatic B cells comprising its fragmentation
`
`(Yamamoto et al. 1981, Morgan et al. 1994). Recent
`experiments have proved that the main reason for the
`STZ-induced B cell death is alkylation of DNA (Delaney
`et al. 1995, Elsner et al. 2000). The alkylating activity of
`STZ is related to its nitrosourea moiety, especially at the
`O6 position of guanine. After STZ injection to rats,
`different methylated purines were found in tissues of
`these animals (Bennett and Pegg 1981).
`Since STZ is a nitric oxide (NO) donor and NO
`was found to bring about the destruction of pancreatic
`islet cells, it was proposed that this molecule contributes
`to STZ-induced DNA damage (Kröncke et al. 1995,
`Morgan et al. 1994). The participation of NO in the
`cytotoxic effect of STZ was confirmed in several
`experiments (Turk et al. 1993, Kröncke et al. 1995).
`Pancreatic B cells exposed to STZ manifested changes
`characteristic for NO action, i.e. increased activity of
`guanylyl cyclase and enhanced formation of cGMP (Turk
`et al. 1993). STZ is, however, not a spontaneous nitric
`oxide donor (Kröncke et al. 1995). This molecule is
`liberated when STZ is metabolized inside cells, but NO
`synthase is not required for this effect (Kröncke et al.
`1995). On
`the other hand,
`the
`lowering of NO
`concentration in pancreatic islet cells by inhibition of the
`inducible
`form of nitric oxide synthase partially
`counteracted DNA cleavage induced by STZ (Bedoya et
`al. 1996). A similar effect can be attained by NO
`scavengers (Kröncke et al. 1995). However, the results of
`several experiments provide the evidence that NO is not
`the only molecule responsible for the cytotoxic effect of
`STZ. STZ was found to generate reactive oxygen species,
`which also contribute to DNA fragmentation and evoke
`other deleterious changes in the cells (Takasu et al.
`1991b, Bedoya et al. 1996). The formation of superoxide
`anions results from both STZ action on mitochondria and
`increased activity of xanthine oxidase (EC 1.1.3.22). It
`was demonstrated that STZ inhibits the Krebs cycle (Turk
`et al. 1993) and substantially decreases oxygen
`consumption by mitochondria (Nukatsuka et al. 1990b).
`These effects
`strongly
`limit mitochondrial ATP
`production and cause depletion of this nucleotide in B
`cells (Nukatsuka et al. 1990b, Sofue et al. 1991).
`Restriction of mitochondrial ATP generation is partially
`mediated by NO. This molecule was found to bind to the
`iron-containing aconitase
`inhibiting enzyme activity
`(Welsh and Sandler 1994).
`Augmented ATP dephosphorylation increases
`the supply of substrate for xanthine oxidase (B cells
`possess high activity of this enzyme) and enhances the
`production of uric acid – the final product of ATP
`
`NOVARTIS EXHIBIT 2090
`Par v. Novartis, IPR 2016-01479
`Page 5 of 10
`
`

`

`542
`
` Szkudelski
`
`Vol. 50
`
`degradation (Nukatsuka et al. 1990a). Then, xanthine
`oxidase catalyses reaction in which the superoxide anion
`is formed (Nukatsuka et al. 1988). As a result of
`superoxide anion generation hydrogen peroxide and
`hydroxyl radicals are formed (Nukatsuka et al. 1990a,
`Takasu et al. 1991b). The inhibition of xanthine oxidase
`by allopurinol restricts the cytotoxic effect of STZ in
`vitro. Pretreatment of B cells with this inhibitor prevented
`the STZ-induced decrease of insulin secretion (Nakatsuka
`et al. 1990a).
`It can be stated that potent alkylating properties
`of STZ are the main reason of its toxicity. However, the
`synergistic action of both NO and reactive oxygen
`species may also contribute to DNA fragmentation and
`other deleterious changes caused by STZ. NO and
`reactive oxygen species can act separately or form the
`highly toxic peroxynitrate (ONOO; Fig. 2). Therefore,
`intracellular antioxidants or NO scavengers substantially
`attenuate STZ toxicity.
`
`Fig. 2. The mechanism of streptozotocin (STZ)-induced
`toxic events
`in B cells of rat pancreas. MIT –
`mitochondria; XOD – xanthine oxidase
`
`References
`
`STZ-induced DNA damage activates poly ADP-
`ribosylation (Sandler and Swenne 1983). This process
`leads to depletion of cellular NAD+, further reduction of
`the ATP content (Heller et al. 1994) and subsequent
`inhibition of insulin synthesis and secretion (Nukatsuka
`et al. 1990b). The concept of unfavorable consequences
`of augmented poly ADP-ribosylation as a result of STZ
`action was confirmed by experiments revealing that the
`inhibition of this process prevents the toxicity of this
`diabetogenic agent. It was found that 3-aminobenzamide,
`a strong
`inhibitor of poly(ADP-ribose)
`synthase,
`protected against the action of STZ in rats, even when
`this substance was administered 45-60 min after STZ
`(Masiello et al. 1985, 1990). Another inhibitor of
`poly(ADP-ribose) synthase, nicotinamide, which is also
`scavenging oxygen free radicals, exerted best protection
`when it was administered shortly after STZ (Masiello et
`al. 1990). The failure of protective action of nicotinamide
`administered after STZ is probably due to a potent
`reduction of the cellular ATP content by STZ since
`nicotinamide uptake is ATP-dependent (Sofue et al.
`1991). The protective effect of 3-aminobenzamide and
`nicotinamide was also confirmed in vitro (Masiello et al.
`1990).
`
`It has been suggested that some inhibitors of
`poly ADP-ribosylation may also exert a protective effect
`due to their hydroxyl radical scavenging properties
`(LeDoux et al. 1988). However, in the case of STZ,
`recent investigations in poly(ADP-ribose) polymerase-
`deficient mice demonstrated that the inhibition of poly
`ADP-ribosylation itself prevents STZ-induced B cell
`damage and hyperglycemia (Pieper et al. 1999). Thus, it
`can be stated that the activation of poly ADP-ribosylation
`is of greater importance for the diabetogenicity of STZ
`than generation of free radicals and DNA damage per se.
`Calcium, which may also induce necrosis, does
`not seem to play a significant role in the necrosis evoked
`by STZ since calcium channel antagonists do not protect
`B cells against streptozotocin, as they do in the case of
`alloxan (Katsumata et al. 1992).
`
`BANSAL R, AHMAD N, KIDWAI JR: Alloxan-glucose interaction: effect of incorporation of 14C-leucine into
`pancreatic islets of rats. Acta Diabetol Lat 17: 135-143, 1980.
`
`NOVARTIS EXHIBIT 2090
`Par v. Novartis, IPR 2016-01479
`Page 6 of 10
`
`

`

`2001
`
`Alloxan and Streptozotocin Action
`
`543
`
`BEDOYA FJ, SOLANO F, LUCAS M: N-monomethyl-arginine and nicotinamide prevent streptozotocin-induced
`double strand DNA break formation in pancreatic rat islets. Experientia 52: 344-347, 1996.
`BENNETT RA, PEGG AE: Alkylation of DNA in rat tissues following administration of streptozotocin. Cancer Res
`41: 2786-2790, 1981.
`BOLAFFI JL, NAGAMATSU S, HARRIS J, GRODSKY GM: Protection by thymidine, an inhibitor of polyadenosine
`diphosphate ribosylation, of streptozotocin inhibition of insulin secretion. Endocrinology 120: 2117-2122,
`1987.
`BOQUIST L, NELSON L, LORENTZON R: Uptake of labelled alloxan in mouse organs and mitochondria in vivo and
`in vitro. Endocrinology 113: 943-948, 1983.
`BOYLAN JM, BRAUTIGAN DL, MADDEN J, RAVEN T, ELLIS L, GRUPPUSO PA: Differential regulation of
`multiple hepatic protein tyrosine phosphatases in alloxan diabetic rats. J Clin Invest 90: 174-179, 1992.
`DEAN PM, MATTHEWS EK: The bioelectrical properties of pancreatic islet cells: effect of diabetogenic agents.
`Diabetologia 8: 173-178, 1972.
`DELANEY CA, DUNGER A, Di MATTEO M, CUNNINGHAM JM, GREEN MH, GREEN IC: Comparison of
`inhibition of glucose-stimulated insulin secretion in rat islets of Langerhans by streptozotocin and methyl and
`ethyl nitrosoureas and methanesulphonates. Lack of correlation with nitric oxide-releasing or O6-alkylating
`ability. Biochem Pharmacol 50: 2015-2020, 1995.
`DONNINI D, ZAMBITO AM, PERRELLA G, AMBESI-IMPIOMBATO FS, CURCIO F: Glucose may induce cell
`death through a free radical-mediated mechanism. Biochem Biophys Res Commun 15: 412-417, 1996.
`DUNN JS, SHEEHAN HL, MCLETHIE NGB: Necrosis of islets of Langerhans produced experimentally. Lancet 1:
`484-487, 1943.
`EBELT H, PESCHKE D, BROMME HJ, MORKE W, BLUME R, PESCHKE E: Influence of melatonin on free
`radical-induced changes in rat pancreatic beta-cells in vitro. J Pineal Res 28: 65-72, 2000.
`EIZIRIK DL, PIPELEERS DG, LING Z, WELSH N, HELLERSTROM C, ANDERSSON A: Major species differences
`between humans and rodents in the susceptibility to pancreatic beta-cell injury. Proc Natl Acad Sci USA 91:
`9253-9256, 1994.
`ELSNER M, GULDBAKKE B, TIEDGE M, MUNDAY R, LENZEN S: Relative importance of transport and
`alkylation for pancreatic beta-cell toxicity of streptozotocin. Diabetologia 43: 1528-1533, 2000.
`GANDA OP, ROSSI AA, LIKE AA: Studies on streptozotocin diabetes. Diabetes 25: 595-603, 1976.
`GIROIX MH, PORTHA B, KERGOAT M, BAILBE D, PICON L: Glucose insensitivity and amino-acid
`hypersensitivity of insulin release in rats with non-insulin-dependent diabetes: a study with the perfused
`pancreas. Diabetes 32: 445-451, 1983.
`GORRAY KC, BASKIN DG, FUJIMOTO WY: Cytotoxic effect of alloxan treatment in vitro on monolayer cultures of
`neonatal rat pancreas. Am J Physiol 254: 417-423, 1983.
`GRANKVIST K: Alloxan-induced luminol luminescence as a tool for investigating mechanisms of radical-mediated
`diabetogenicity. Biochem J 200: 685-690, 1981.
`GRANKVIST K, MARKLUND S, SEHLIN J, TÄLJEDAL IB: Superoxide dismutase, catalase and scavengers of
`hydroxyl radical protect against the toxic action of alloxan on pancreatic islet cells in vitro. Biochem J 182: 17-
`25, 1979.
`GRUPPUSO PA, BOYLAN JM, POSNER BI, FAURE R, BRAUTIGAN DL: Hepatic protein phosphotyrosine
`phosphatase. Dephosphorylation of insulin receptor and epidermal growth factor receptors in normal and
`alloxan diabetic rats. J Clin Invest 85: 1754-1760, 1990.
`HARMAN AW, FISCHER LJ: Alloxan toxicity in isolated rat hepatocytes and protection by sugars. Biochem
`Pharmacol 31: 3731-3736, 1982.
`HEIKKILA RE, WINSTON B, COHEN G, BARDEN H: Alloxan induced diabetes, evidence for hydroxyl radicals as a
`cytotoxic intermediate. Biochem Pharmacol 25: 1085-1092, 1976.
`HELLER B, BURKLE A, RADONS J, FENGLER E, JALOWY A, MULLER M, BURKART V, KOLB H: Analysis
`of oxygen radical toxicity in pancreatic islets at the single cell level. Biol Chem Hoppe-Seyler 375: 597-602,
`1994.
`
`NOVARTIS EXHIBIT 2090
`Par v. Novartis, IPR 2016-01479
`Page 7 of 10
`
`

`

`544
`
` Szkudelski
`
`Vol. 50
`
`JÖRNS A, MUNDAY R, TIEDGE M, LENZEN S: Comparative toxicity of alloxan, N-alkylalloxans and ninhydrin to
`isolated pancreatic islets in vitro. J Endocrinol 155: 283-293, 1997.
`JÖRNS A, TIEDGE M, LENZEN S, MUNDAY R: Effect of superoxide dismutase, catalase, chelating agents, and free
`radical scavengers on the toxicity of alloxan to isolated pancreatic islets in vitro. Free Radic Biol Med 26:
`1300-1304, 1999.
`KANDULSKA K, SZKUDELSKI T, NOGOWSKI L: Lipolysis induced by alloxan in rat adipocytes is not inhibited by
`insulin. Physiol Res 48: 113-117, 1999.
`KATSUMATA K, KATSUMATA K, Jr., KATSUMATA Y: Protective effect of diltiazem hydrochloride on the
`occurrence of alloxan- or streptozotocin-induced diabetes in rats. Horm Metab Res 24: 508-510, 1992.
`KATSUMATA K, KATSUMATA Y, OZAWA T, KATSUMATA K, Jr.: Potentiating effects of combined usage of
`three sulfonylurea drugs on the occurrence of alloxan diabetes in rats. Horm Metab Res 25: 125-126, 1993.
`KIM HR, RHO HW, PARK JW, KIM JS, KIM UH, CHUNG MY: Role of Ca2+ in alloxan-induced pancreatic beta-cell
`damage. Biochim Biophys Acta 1227: 87-91, 1994.
`KLIBER A, SZKUDELSKI T, CHICHLOWSKA J: Alloxan stimulation and subsequent inhibition of insulin release
`from in situ perfused rat pancreas. J Physiol Pharmacol 47: 321-328, 1996.
`KRÖNCKE KD, FEHSEL K, SOMMER A, RODRIGUEZ ML, KOLB-BACHOFEN V: Nitric oxide generation during
`cellular metabolization of the diabetogenic N-methyl-N-nitroso-urea streptozotocin contributes to islet cell
`DNA damage. Biol Chem Hoppe-Seyler 376: 179-185, 1995.
`I, SAKAMOTO K: Effect of
`KURAHASHI M, MASUI H, YOSHIMOTO S, WAKABAYASHI
`diethyldithiocarbamate on diabetogenic action of alloxan in rats. Diabetes Res Clin Pract 19: 39-47, 1993.
`LEDOUX SP, HALL CR, FORBES PM, PATTON NJ, WILSON GL: Mechanisms of nicotinamide and thymidine
`protection from alloxan and streptozotocin toxicity. Diabetes 37: 1015-1019, 1988.
`LENZEN S, MIRZAIE-PETRI M: Inhibition of glucokinase and hexokinase from pancreatic B-cells and liver by
`alloxan, alloxantin, dialuric acid, and t-butylhydroperoxide. Biomed Res 12: 297-307, 1991.
`LENZEN S, MUNDAY R: Thiol-group reactivity, hydrophilicity and stability of alloxan, its reduction products and its
`N-methyl derivatives and a comparison with ninhydrin. Biochem Pharmacol 42: 1385-1391, 1991.
`LENZEN S, PANTEN U: Alloxan: history and mechanism of action. Diabetologia 31: 337-342, 1988.
`LENZEN S, TIEDGE M, PANTEN U: Glucokinase in pancreatic B-cells and its inhibition by alloxan. Acta Endocrinol
`(Copenh) 115: 21-29, 1987.
`LENZEN S, FREYTAG S, PANTEN U: Inhibition of glucokinase by alloxan through interaction with sugar-binding
`site of the enzyme. Mol Pharmacol 34: 395-400, 1988.
`LENZEN S, BRÜNIG H, MÜNSTER W: Effects of alloxan and ninhydrin on mitochondrial Ca2+ transport. Mol Cell
`Biochem 118: 141-151, 1992.
`LENZEN S, TIEDGE M, JORNS A, MUNDAY R: Alloxan derivatives as a tool for the elucidation of the mechanism
`of the diabetogenic action of alloxan. In: Lessons from Animal Diabetes, E SHAFRIR (ed), Birkhauser,
`Boston, 1996, pp 113-122.
`MALAISSE WJ, MALAISSE-LAGAE F, SENER A, PIPELEERS DG: Determinants of the selective toxicity of
`alloxan to the pancreatic B cell. Proc Natl Acad Sci USA 79: 927-930, 1982.
`MALAISSE WJ: Alloxan toxicity to the pancr

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket