WO2009026317A2 - Use of eif-5a1 sirna to protect islets cells from apoptosis and to preserve their functionality - Google Patents

Use of eif-5a1 sirna to protect islets cells from apoptosis and to preserve their functionality Download PDF

Info

Publication number
WO2009026317A2
WO2009026317A2 PCT/US2008/073642 US2008073642W WO2009026317A2 WO 2009026317 A2 WO2009026317 A2 WO 2009026317A2 US 2008073642 W US2008073642 W US 2008073642W WO 2009026317 A2 WO2009026317 A2 WO 2009026317A2
Authority
WO
WIPO (PCT)
Prior art keywords
eif
sirna
islet
islets
cells
Prior art date
Application number
PCT/US2008/073642
Other languages
French (fr)
Other versions
WO2009026317A3 (en
Inventor
John E. Thompson
Charles A. Dinarello
Original Assignee
Senesco Technologies, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Senesco Technologies, Inc. filed Critical Senesco Technologies, Inc.
Priority to AU2008288988A priority Critical patent/AU2008288988A1/en
Priority to JP2010521983A priority patent/JP2010536379A/en
Priority to EP08798216A priority patent/EP2195429A2/en
Priority to CN2008801121650A priority patent/CN102124108A/en
Priority to CA2700463A priority patent/CA2700463A1/en
Publication of WO2009026317A2 publication Critical patent/WO2009026317A2/en
Publication of WO2009026317A3 publication Critical patent/WO2009026317A3/en
Priority to IL204072A priority patent/IL204072A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0676Pancreatic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/48Regulators of apoptosis

Definitions

  • the islets of Langerhans is a multi-cellular entity containing cells that produce insulin within the pancreas.
  • the average person has about a million islets, and they contain approximately two to three percent of the total number of cells in the pancreas.
  • the pancreas contains the islets of Langerhans, which house beta cells that produce insulin.
  • the beta cells monitor glucose levels in the blood and release finely measured amounts of insulin to counterbalance glucose peaks. Type I and II diabetes develop when more than 90 percent of these beta cells are damaged.
  • Type 1 diabetes is a disorder of glucose homeostasis that affects -10% of the 21 million diabetic individuals in the USA. Type 1 diabetes results from the virtually complete autoimmune destruction of pancreatic islet ⁇ cells, leaving individuals dependent upon the administration of insulin to maintain life. The pathogenesis of Type 1 diabetes involves a complex interplay between cells of the immune system and antigens present on the islet ⁇ cell.
  • ThI cells and macrophages which subsequently release, among other factors, cytokines (ILl ⁇ , TNF ⁇ , and IFN ⁇ ) that induce nitric oxide production, which triggers both apoptotic and necrotic islet ⁇ cell death
  • cytokines ILl ⁇ , TNF ⁇ , and IFN ⁇
  • pancreas or islet transplantations have been variably successful in reversing type 1 diabetes, but require tissues from limiting sources (cadaveric donors) and are still subject to destruction by the underlying autoimmune process.
  • Islet transplantation is a most promising and minimally physiologically invasive procedure for treatment of Type I diabetes mellitus. Transplanting islets rather than complete pancreatic tissue has the distinct advantages of ease of transplantation, and the elimination of the pancreatic exocrine function of the donor tissue involving secretion of digestive en2ymes. Liberating islets from pancreatic exocrine tissue is the initial and crucial step that influences islet transplantations. The important objective in islet isolation is to provide sufficient numbers of viable functional and potent islets for transplantation. The "Edmonton Protocol" transplants healthy islets into diabetic patients.
  • Islet transplantation using the Edmonton Protocol is described in Shapiro, Ryan, and Lakey, Clinical Islet Transplantation-State of the Art, Transplantation Proceedings, 33, pp. 3502-3503 (2001); Ryan et al., Clinical Outcomes and Insulin Secretion After Islet Transplantation With the Edmonton Protocol, Diabetes, Vol. 50, April 2001, pp. 710-719; and Ryan elal., Continued Insulin Reserve Provides Long-Term Glycemic Control, Diabetes, Vol. 51, July 2002, pp. 2148- 2157. Once in the liver, the cells develop a blood supply and begin producing insulin.
  • the Edmonton Protocol may include 7-10 steps depending on the method employed.
  • the first step involves the delivery of a specific enzyme (liberase) to a donor pancreas, which digests the pancreas tissue, but does not digest the islets.
  • a specific enzyme liberase
  • the separated islets are transplanted into the main vessel of the liver, known as the portal vein.
  • the liver is able to regenerate itself when damaged, building new blood vessels and supporting tissue. Therefore, when islets are transplanted into the liver, it is believed that new blood vessels form to support the islets.
  • the insulin that the cells produce is absorbed into the blood stream through these surrounding vessels and distributed through the body to control glucose levels in the blood.
  • Islet transplantation together with steroid-free, nondiabetogenic immunosuppressive therapy, has been used to treat patients with Type 1 diabetes.
  • Such treatments can lead to increased risk of hyperlipidemia and hypertension, and long-term studies demonstrate that islet viability is impaired.
  • Edmonton protocol of islet transplantation in humans has demonstrated remarkable short term success, with 80% of individuals achieving insulin independence at 1 year post- transplant. However, this rate decreases to only about 10-15% at 5 years.
  • the reasons for this progressive loss of graft function is unclear, but likely includes islet loss as a result of ongoing cytokine-mediated inflammation.
  • islet transplantation may continue to remain a viable therapeutic option for individuals with Type 1 diabetes, particularly as results show that even those that fail long-term insulin independence, they still retain other important benefits, such as decreased blood sugar lability, lower incidence of hypoglycemia, and decreased insulin dosage.
  • the first days following islet transplantation are a particularly vulnerable period for the transplanted islets.
  • the present invention provides a method for preserving the functionality of harvested islet cells after isolation comprising administering eIF-5Al siRNA to the islet cells of an islet cell donor prior to islet isolation, wherein the eIF-5Al siRNA inhibits expression of eIF-5Al in the islet cells and thereby inhibits apoptosis in the islet cells and preserves the functionality of the harvested islet cells.
  • the siRNA targets the following nucleotide sequences of eIF-5Al: 5'-AAAGGAATGACTTCCAGCTGA-S'; 5'- AAGATCGTCGAGATGTCTACT-3'; 5'-AAGGTCCATCTGGTTGGTATT-S'; or 5'- AAGCTGGACTCCTCCTACACA-3'.
  • the eIF-5Al siRNA comprises the nucleotide sequence 5'-AAAGGAAUGACUUCCAGCTGAdTdT-S'.
  • the siRNA may be administered to the donor through any acceptable means. In certain embodiments, the siRNA is administered to the islet cell donor via intraperotoneal injection.
  • Another embodiment of the present invention provides a method for inhibiting islet cells from undergoing apoptosis during a donor harvesting process comprising administering eIF-5Al siRNA to an islet cell donor via intraperotoneal injection to islet cell donor prior to islet isolation, wherein the eIF-5Al siRNA inhibits expression of eIF-5Al in the islet cells and thereby inhibits apoptosis in the islet cells.
  • the siRNA targets the following nucleotide sequences of elF- 5Al: 5'-AAAGGAATGACTTCCAGCTGA-S'; 5'-AAGATCGTCGAGATGTCTACT-S'; 5'- AAGGTCCATCTGGTTGGTATT-S'; or 5'-AAGCTGGACTCCTCCTACACA-S'.
  • the eIF-5Al siRNA comprises the nucleotide sequence 5'- AAAGGAAUGACUUCCAGCTGAdTdT-3'.
  • siRNA or antisense construct can be used, as long as such construct inhibits expression of eIF-5Al in the islet cells.
  • Administration of siRNA may be by any suitable route.
  • Exemplary administration methods include perfusion through the portal vein of the islet cell donor, hydrodynamic perfusion through the portal vein of the islet cell donor, and intraperotoneal administration.
  • the present invention also provides a composition for inhibiting apoptosis and preserving functionality of in islet cells, comprising eIF-5Al siRNA, wherein the siRNA inhibits expression of eIF-5Al and thereby inhibits apoptosis in the islet cells.
  • eIF-5Al siRNAs are discussed above.
  • the present invention also provides a method of inhibiting expression of a target protein in islet ⁇ cells comprising administering to a ⁇ cell donor prior to ⁇ cell harvesting an siRNA construct targeted to the mRNA encoding the target protein, wherein the siRNA inhibits expression of the target protein in the islet ⁇ cells.
  • the invention also provides a method of decreasing the production of nitric oxide (NO) via the inducible nitric oxide synthase (iNOS) pathway, by decreasing expression of eIF-FA.
  • NO nitric oxide
  • iNOS inducible nitric oxide synthase
  • the invention also provide the use of eIF5A siRNA to manufacture a medicament to decrease production of nitric oxide via the iNOS pathway.
  • Figure 1 provides results of RT-PCR performed for ⁇ -actin, mAAT and eIF-5Al after perfusion through the portal vein with eIF-5A siRNA. This figure shows that eIF-5Al expression is measurable and was thus incorporated into islets.
  • Figure 2 shows slows retrograde portal vein perfusion. Bile duct (clear) and portal vein (red) ready for preparatory knot (dark suture). The needle enters below the knot (direction indicated by arrow), cross under the knot and releases siRNA into vessels that reach pancreas, spleen, intestine and a third of distal colon.
  • Figure 3 shows that perfusion of eIF-5Al siRNA into islets causes a reduction of expression of eIF-5Al (shown is reduction in mRNA levels of eIF-5Al).
  • Figure 6 provides the nucleotide sequence of human eIF-5Al aligned against eIF5-A2.
  • Figure 7 provides the amino acid sequence of human eIF-5Al aligned against eIF5-A2.
  • Figure 8 provides the nucleotide sequence of human eIF-5Al with exemplary antisense oligonucleotides.
  • Figure 9 provides the nucleotide sequence of human eIF-5Al with exemplary antisense oligonucleotides.
  • Figures 1OA and B provide the nucleotide sequence of human eIF-5Al with exemplary siRNAs.
  • Figure 11 provides the nucleotide sequence of human eIF-5Al with exemplary siRNAs.
  • Figure 12 shows a schematic of experiments showing that siRNA eIF-5Al was able to knock down (decrease) expression in islet cells.
  • Figure 13A shows the pattern of insulin secretion for 50 islets and Figure 13B shows the pattern of [Ca2+]I from a single islet following 2.8 to 11 mM glucose stimulation. The phases are by 0, 1, 2.
  • This figure shows a normally functioning islet responds to glucose stimulation with a biphasic pattern of insulin secretion, which closely resembles similar changes in islet intracellular calcium ([Ca 2+ ];).
  • Figure 14 provides the results of calcium oscillation studies. These figures show that islet cells treated with eIF-5Al siRNA (and thus have decreased expression of eIF-5Al) showed a significantly stronger glucose response and overall oscillation. This indicates that these treated cells not only survive longer but also retain functionality.
  • Figure 15 shows different photographs of stained cytokine treated islet cells.
  • the photographs shows that islet cells treated with eIF-5Al siRNA have more live and less dead cells than the control or vehicle treated (saline only) islet cells.
  • Figures 16A and 16B show through four different imaging techniques that the siRNA gets into islets cells when administered to the islet cell donor via IP injection. Confocal images were used to generate 3D reconstructions of isolated islets.
  • Figure 16A shows that islets isolated from mice administered 0.9% saline intraperitoneally once daily for three days show characteristic FITC autofluorescence but minimal fluorescence in the TRITC channel (labeled
  • FIG. 16B shows that islets isolated from C57BI/6 mice administered Cy3- labeled stabilized siRNA intraperitoneally once daily for three days show penetrance of the Cy3 label.
  • Figure 17 (A) shows the results of where ⁇ TC3 cells were transfected with two different siRNA constructs (constructs A and B) or controls and subjected to immunoblotting for the indicated proteins.
  • Figure 17 (B) shows the results of where C57B1/6 mice were injected with the same siRNAs intraperitoneally once daily for three days, and islets were isolated and purified via collagenase digestion and differential gradient centrifugation. Islets were lysed in 2% SDS and subjected to immunoblotting.
  • Figure 17 (C) shows the results where isolated islets were loaded with Fura2 for 30 minutes before imaging in 3mM D-glucose.
  • FIG. 17(D) shows the results where glucose stimulated insulin secretion (GSIS) was performed using 50 islets from each treatment group.
  • Figures 18A-I show that knockdown of eIF5A improvise islet function and survival ex- vivo.
  • Figures 19A-D show that cytokine induced of iNOS protein production is absent in eIF5A-deficient cells.
  • Eukaryotic initiation factor 5A appears to be a crucial factor in the post- transcriptional regulation of stress-induced genes, and appears to promote cytokine-mediated apoptosis in mammalian cells.
  • eIF5A is a small acidic protein that is very highly conserved throughout eukaryotes, and is the only protein known to contain the unique polyamine-derived amino acid hypusine.
  • eIF5 A has been proposed to play roles in mRNA processing, trafficking, and translation. However, its role, if any, in the inflammatory cascade in pancreatic islets has not before been characterized. Pancreatic islets are highly vulnerable to glucose and lipid toxicity, oxidative stress, and inflammatory cytokines.
  • RNA interference small interfering RNAs
  • RNA interference studies in primary rodent islets are their relatively poor transfection efficiency.
  • this technique is time-consuming to generate the appropriate viruses and is subject to viral toxicity.
  • the inventors discovered a novel protocol to deplete mouse islets of selected proteins in vivo by repeated intraperitoneal injections of stabilized small interfering RNAs (siRNAs). This protocol results in remarkable penetration of siRNAs within the islet.
  • This technique can be used to successfully deplete islets of any desired/target protein such as the well-characterized pancreatic transcription factor Pdxl .
  • eIF5A contributes to cytokine-mediated islet dysfunction by promoting translation of the mRNA encoding inducible nitric oxide synthase (iNOS).
  • iNOS inducible nitric oxide synthase
  • eIF5A eukaryotic initiation factor 5A
  • eIF5A eukaryotic initiation factor 5A
  • eIF5A eukaryotic initiation factor 5A
  • eIF5A has been characterized in other systems as a crucial regulator in the translation of stress-induced genes.
  • siRNA against eIF5A was injected into mice, a 50-70% decrease in eIF5A protein levels in isolated islets was observed. Accompanying this fall, a 30-40% increase in glucose stimulated insulin secretion and Ca2+ mobilization as compared to control treatment was observed.
  • isolated islets were exposed to a cocktail of proinflammatory cytokines (ILl ⁇ , TNF ⁇ , IFN ⁇ ), the relative enhancement of islet function persisted in animals treated with si-eIF5A.
  • proinflammatory cytokines ILl ⁇ , TNF ⁇ , IFN ⁇
  • Islet protection following eIF5A knockdown was not accompanied by alterations in the expression of genes whose products are involved in glucose responsiveness or insulin transcription (Slc2a2, Gck, Irsl, Nkx6-1, MafA, Pdxl, NeuroDl, and Setd7). See Figure 18C.
  • iNOS inducible nitric oxide synthase
  • eIF5A plays an essential role in islet ⁇ cell response to stress signals via control of iNOS translation, and as such, eIF5A may serve as a viable target for therapeutic strategies aimed at preserving viability as well as the ability of the islet cells to function after harvesting and post implantation.
  • siRNA incorporation into islets can be achieved by pancreatic perfusion via retrograde portal vein inoculation. See Bradley, et al., Transplantation Proceedings, 37, 233-236, 2005. Briefly, Cy-3 labeled Luciferase (Luc) siRNA GL2 duplex was used either packaged with Lipofectamine 2000 or unpackaged, and injected either through tail vein (in vivo, 50 ⁇ g per mouse) or directly into the pancreas by retrograde portal vein inoculation (in situ, 2 ⁇ g per mouse). Pancreata were procured and stored at 4 0 C for 24 hours after in situ delivery, or 4 hours after in vivo delivery, and islets were isolated and cultured an extra 16 hours before examination.
  • Figure 1 shows that perfusion to the islet cells provides a suitable delivery mechanism to the islet cells.
  • the present invention provides a method for inhibiting expression of a target protein in islet cells comprising administering siRNA targeted to the mRNA encoding the target protein to the islet cells, wherein the siRNA inhibits expression of the target protein in the islet cells.
  • Administration may be through any means, preferably trough intraperitoneal administration to the islet ⁇ cell donor before the islet ⁇ cell isolation.
  • the present invention provides a method for inhibiting expression of eIF-5Al in islet cells comprising administering eIF-5Al siRNA to the islet cells, wherein the eIF-5Al siRNA inhibits expression of eIF-5Al in the islet cells.
  • Figures 16A and 16B show that administration to the islet cell donor via IP injection also provides a suitable delivery mechanism to the islet cells.
  • Figure 16B shows that three daily intraperitoneal injections of stabilized, Cy3-labeled double-stranded RNAs into C57BL/6 mice revealed remarkable penetration into isolated islets, as determined by confocal microscopy.
  • Figures 12B and 18B show that the eIF-5Al siRNA treated islet cells do indeed express less eIF-5Al siRNA.
  • the present invention also provides a method for inhibiting apoptosis in harvested islet cells comprising administering eIF-5 Al siRNA to the islet ⁇ cell donor, wherein the elF- 5Al siRNA inhibits expression of eIF-5Al in the islet cells and wherein the inhibition of elF- 5Al expression inhibits apoptosis.
  • the islets cells are treated with eIF-5Al siRNA cells prior to the harvesting process.
  • the present invention further provides a method for preserving the functionality of harvested islet cells after isolation comprising administering eIF-5Al siRNA to the islet cells of an islet cell donor prior to islet isolation, which results in a inhibition or reduction of eIF-5Al expression in the islet cells, which in turn inhibits apoptosis in the islet cells, as well as preserves their functionality.
  • Preserving the functionality of the islet cells means that not only do the islet cells survive the harvesting process, but also maintain their function (the functionality of the cells remains longer and/or they maintain a better glucose response than non eIF-5Al siRNA treated islet cells)(i.e. release insulin in response to glucose) after the harvesting process.
  • IP intraperitoneal
  • Protein lysates from each group of islets were analyzed by Western Blot for protein knockdown.
  • Figure 12B illustrates the relative knockdown of eIF-5Al in Group III treated mouse islets, which is quantified in Figure 12C.
  • Figure 12C illustrate the efficacy of IP injection to produce knockdown expression of eIF-5Al in islets.
  • Evidence of preserved functionality of the harvested islets is demonstrated by calcium oscillation assays. The inventors have determined that intracellular calcium is an indicator of stimulation and stress. For both humans and rodents, a normally functioning islet responds to glucose stimulation with a biphasic pattern of insulin secretion, as shown in Figure 13 A.
  • this biphasic secretion pattern closely resembles similar changes in islet intracellular calcium ([Ca 2+ ]O, which also consists of a first phase peak that drops into a second phase plateau ( Figure 13B).
  • the [Ca 2+ ]; response to glucose consists of three phases (see figure 13) that roughly reflects the processes described by the "Consensus Model" of beta-cell stimulus- secretion coupling.
  • Disruptions in glucose metabolism, mitochondria and ATP production, endoplasmic reticulum (ER), ion channel function, and myriad other problems can affect the dynamics of calcium handling related to glucose stimulation.
  • ER endoplasmic reticulum
  • defects in these aspects of activity are more likely to be observed than by standard measures of static insulin secretion. For example, increased basal [Ca 2+ J 1 in low glucose and the loss of the phase 0 dip in [Ca 2+ J; during glucose stimulation can be indicative of ER-stress or possibly ion-channel dysfunction.
  • Such defects can not be detected easily by measuring static or kinetic insulin secretion, but the imaging techniques described herein can be utilized to address these questions.
  • the present invention further provides a method of inhibiting apoptosis in cells involved in inflammtory diseases in which the disease is associated with increased production of nitric oxide.
  • inflammatory diseases such as rheumatoid arthritis (RA) are associated with increased production of nitric oxide (NO), due to activation of the inducible nitric oxide synthase (iNOS) pathway.
  • NO nitric oxide
  • iNOS inducible nitric oxide synthase
  • synovial fibroblasts Several cell types present within the joint, including synovial fibroblasts, endothelial cells and chondrocytes, can be induced by pro- inflammatory cytokines to produce NO in vitro.
  • localization studies have shown that there is up-regulation of iNOS expression in synovial lining cells, chondrocytes and blood vessels in joint tissues obtained from patients with RA.
  • the localization of iNOS expression to the synovial lining layer and cartilage is of interest in the light of other studies that have shown that apoptosis is increased in RA, particularly in the synovial lining layer and cartilage.
  • the present invention provides a method of inhibiting apoptosis in cells associated with RA (such as synovial and cartilage cells) by inhibiting expression of eIF5A and hence inhibits or decreases the production of nitric oxide via the iNOS pathway to inhibit apoptosis in the cells.
  • the present invention further provides a method of inhibiting NO production in a cell or host by administration to the cell or host an siRNA directed against eIF-5A.
  • the siRNA decreases expression of eIF-5A and in turn decreases the production of NO by inhibiting the activation of the iNOS pathway.
  • the invention also provide the use of eIF5 A siRNA to manufacture a medicament to decrease production of nitric oxide via the iNOS pathway.
  • any eIF-5Al siRNA that inhibits expression of eIF-5Al may be used.
  • the term “inhibits” also means reduce as compared to levels that would occur in control cells, i.e. those cells not having been treated with eIF-5Al siRNA.
  • One exemplary and especially preferred elF- 5Al siRNA comprises the sequence: 5'-AAAGGAAUGACUUCCAGCTGAdTdT-S'.
  • Co- pending application 11/293,391 which was filed on November 28, 2005 (which is herein incorporated by reference in its entirety) provides additional exemplary eIF-5Al siRNAs and other antisense constructs that have been used to inhibit expression of eIF-5Al in other cell types and were also shown to inhibit apoptosis.
  • Figures 6-11 provide sequences of eIF-5Al, exemplary eIF-5Al siRNAs and antisense constructs.
  • antisense constructs of eIF-5Al may be used to inhibit expression of eIF-5Al and thus inhibit apoptosis of the islet cells, as well as maintain or preserve their functionality.
  • the eIF-5Al siRNA targets the following nucleotide sequence ofeIF-5Al (see figure 10): 5'-AAAGGAATGACTTCCAGCTGA-S'; 5'-AAGATCGTCGAGATGTCTACT-S'; 5'-AAGGTCCATCTGGTTGGTATT-S'; and 5'- AAGCTGGACTCCTCCTACACA-3'.
  • the siRNA targets the following sequence of eIF-5Al: 5'-AAAGGAATGACTTCCAGCTGA-S'.
  • the present invention also provides a method for inhibiting islet cells from undergoing apoptosis during a donor harvesting process.
  • many islets cells undergo apoptosis when they are harvested.
  • the present inventors have shown that providing eIF-5Al siRNA to the islet cells prior to harvesting, offers a protective benefit against apoptosis.
  • the eIF-5Al siRNA is administered to the islet cells of an islet cell donor prior to islet isolation.
  • the donor and hence islet cells
  • the donor may be any animal, including human islet cells. Any method of administration may be used.
  • the siRNA may be administered via perfusion through the portal vein of the islet cell donor or via hydrodynamic perfusion through the portal vein of the islet cell donor. See example 1.
  • Another form of administration includes intraperotoneal administration. See example 2 and examples 3-5.
  • Perfusion through portal vein is similar to canulation of the bile duct, but the needle points the opposite way.
  • the portal vein is exposed by retraction of liver and shifting of visceral organs to the mouse's left.
  • a preparative knot is made around it and includes the bile duct. After puncturing the vessel a blunted needle is advanced toward the pancreas and the knot is tightened around it.
  • 1 ml saline or siRNA 5 ⁇ g
  • the needle is removed and the knot is closed behind the needle to prevent fluid escape.
  • the pancreas may be held longer with siRNA. Alternatively, it can be removed but kept cold with collagenase longer.
  • Regular islet isolation methods are followed and the islets (50) may be incubated in for 16 hours.
  • the present invention also provides a composition for inhibiting apoptosis in islet cells, comprising eIF-5Al siRNA, wherein the siRNA inhibits expression of eIF-5Al and thereby inhibits apoptosis and maintains the functionality of the islet cells.
  • the composition may comprise other or additional eIF-5Al siRNAs as discussed above.
  • a preferred siRNA comprises the nucleotide sequence 5'-AAAGGAAUGACUUCCAGCTGAdTdT-S' EXAMPLES
  • Example 1 Portal vein perfusion
  • Mouse islets express elFSA.
  • Total RNA was extracted from isolated mouse islets and RT-PCR was performed for ( ⁇ -actin and for eIF-5Al (fig. 1). Resting non-stimulated islets exhibited positive levels of eIF-5Al-mRNA.
  • elFSAl-mRNA levels diminished after elF5Al-siRNA delivery: portal vein slow perfusion.
  • Pancreata were digested by collagenase irrigation of pancreatic duct and islets were isolated as described by
  • eIF-5Al-mRNA levels diminished and islet apoptosis rate reduced after elF5A-siRNA delivery: portal vein hydrodynamic perfusion.
  • Pancreata were digested by collagenase irrigation of pancreatic duct and islets were isolated. Islets were incubated for 16 hours and then divided: one group was stained with propidium iodide for evaluation of apoptosis (50 islets per mouse) and the other group was processed for RT-PCR (25 islets per mouse).
  • siRNA molecules were synthesized by Dhaimacon, Lafayette, CO. The sequence of the eIF-5Al and control siRNA were: 5' CGGAAUGACUUCCAGCUGAdTdT 3 1 and 5' AGUCGACCUUCAGUAAGGCdTdT 3', respectively.
  • eIF-5Al Primers Forward 5'-GAC AGT GGG GAG GTA CGA GA-3'; Reverse 5'-GGG GTG AGG AAA ACC AAA AT-3 1 .
  • PI Propidium iodide
  • Antibodies and siRNA Mouse monoclonal antibody against eIF-5Al were generated. Anti-actin monoclonal antibody (clone C4, #69100) was purchased from MP Biomedicals. In Western blots, near infrared fluorophore labeled secondary antibodies from Li-Cor were used (IRDye 800 and IRDye 700). The siRNA specific for eIF-5Al was directed to the following sequence of eIF-5Al : 5'-AAAGGAATGACTTCCAGCTGA-S'. While control siRNA was the transcribed sequence, 5'-AAAGTCGACCTTCAGTAAGGA-S', and was previously shown to not induce knockdown of any known protein. See co-pending applications U.S. 11/725,470 and PCT/US07/64424, herein incorporated by reference in their entireties.
  • IP (intraperotoneal) Injection of mice with siRNA Twelve, 8- to 10-week old C573L/6 male mice were purchased from Charles River. Male mice were chosen to avoid the metabolic variation that accompanies the female estrous cycle. Mice were randomly assigned one of three treatment groups: Group I - vehicle treatment (0.9% saline), Group II - control siRNA treatment, Group III - eIF-5Al siRNA treatment. Each mouse received the select treatment via IP injection with 16.6mg/kg or similar volume (in the case of Group I). Mice were injected each day (approximately 11 :00 a.m.) for three days (Days -3, -2, -1) prior to islet harvest (Day 0). Islet Isolations and Cytokine Treatment: Islet isolations from C57BL/6 mice were carried out using standard isolation techniques and a protocol approved by the Institutional
  • Pancreata were stored in HBSS supplemented with 4.2mM sodium bicarbonate and 1% BSA (Invitrogen) until collagenase digestion. Purification was performed by differential centrifugation. Whole islets were maintained in phenolred-free DMEM solution supplemented with 10% FBS and 5% penicillinn/streptomycin. Cultures were incubated at 37 0 C + 5% CO 2 in 5.5mM glucose. Purified islets were allowed to recover for 16- 18 hours before metabolic testing (calcium oscillations).
  • Cytokine treatment was applied to half of the islets at the end of metabolic testing on Day 1 using a prescribed cocktail of physiologically relevant cytokines (5ng/mL IL l ⁇ , lOng/mL TNT- ⁇ , 100ng/mL INF- ⁇ . Cytokine treated islets were allowed to rest 24 hours before further testing and protein analysis commenced (Day 2 and Day 3). Immunoblot Assays: 10 ⁇ g of islet cell extract (prepared in Laemmli buffer containing
  • Example 3 Application of siRNA against Pdx-1 in vitro and in vivo ⁇ TC3 cells were transfected with two different Pdx-1 siRNA constructs (constructs A and B) or controls and subjected to immunoblotting for the proteins Pdx-1, GAPDH and ⁇ -actin.
  • constructs A and B constructs A and B
  • islets were isolated and purified via collagenase digestion and differential gradient centrifugation. Islets were lysed in 2% SDS and subjected to immunoblotting.
  • Figure 17B Isolated islets were loaded with Fura2 for 30 minutes before imaging in 3mM D- glucose.
  • Islets were stimulated with 1 ImM D-glucose at ⁇ 300 seconds, and the Fura2 ratio was continuously monitored by fluorescence microscopy. See Figure 17C.
  • Glucose stimulated insulin secretion (GSIS) was performed using 50 islets from each treatment group. See Figure 17D.
  • Example 4 Knockdown of eIF-5A improves islet function and survival ex vivo
  • FIG. 18A provides the schematic representation of experimental design. Islets were isolated post-treatment and extracts were subjected to immunoblot analysis. See figure 18B. Isolated islets were subjected to RT-PCR analysis of genes essential to glucose sensing and insulin transcription; although gene transcription was inhibited by cytokine treatment (4 h incubation with IFNg, ILIb, TNFa), no differences between groups were observed. See Figure 18C.
  • si-eIF5A-treated islets 24h (see figure 18D) and 48h (see figure 18F) after isolation show improved Glucose Stimulated Calcium responses (GSCa) compared to controls at 24 h (see figure 18D) and 48 h (see figure 18F) after isolatin.
  • si-eIF5A-treated islets demonstrate enhanced GSCa in the presence of cytokines 24h (see figure 18E) and 48h (see figure 18G) after isolation.
  • Islets isolated from eIF5A treated mice showed improved glucose stimulated insulin secretion ("GSIS") compared to controls in both the absence (see figure 18H) and presence (see figure 18H) of 4h cytokine incubation, as assessed by GSIS.
  • GSIS glucose stimulated insulin secretion
  • Example 5 Knockdown of eIF-5A abrogates cytokine-induced INOS production
  • Cytokine induced of iNOS protein production is absent in eIF5A-deficient cells. Islets treated with cytokines were subjected to RT-PCR for iNOS mRNA; all treatment groups showed dramatic upregulation of iNOS mRNA upon cytokine exposure. See figure 19A. Cytokine- treated islets were subjected to immunoblot analysis. Notably, iNOS protein was induced upon cytokine exposure in control islets, but not si-eIF5A-treated islets. See Figure 19B. INS-I (832/13) ⁇ cell were treated with cytokine cocktail for 4 h and subjected to immunoblotting; cells show similar increases in iNOS protein. See figure 19C.
  • INS-I cells were treated with varying concentrations of GC-7, an inhibitor of deoxyhypusine synthase, overnight, then incubated for 4h in the presence of cytokines.
  • INS-I cells show an inverse correlation of iNOS production and GC-7 concentration, suggesting that inhibition of active eIF5A production prevents iNOS translation. See figure 19D.
  • Turpaev KT Amchenkova AM
  • Narovlyansky AN Two pathways of the nitric oxide- induced cytotoxic action. Biochem MoI Biol Intl997;41:1025-33.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Diabetes (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Cell Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Emergency Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Endocrinology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Agricultural Chemicals And Associated Chemicals (AREA)

Abstract

The present invention relates to methods for improving the viability, recovery and functionality of islets that are separated from a donor organ for subsequent transplantation and more particularly relates to the use of eIF-5A1 siRNAs to enhance the viability and functionality of islets.

Description

USE OF EIF-5A1 SIRNA TO PROTECT ISLETS CELLS FROM APOPTOSIS AND TO PRESERVE THEIR FUNCTIONALITY
BACKGROUND OF THE INVENTION The islets of Langerhans is a multi-cellular entity containing cells that produce insulin within the pancreas. The average person has about a million islets, and they contain approximately two to three percent of the total number of cells in the pancreas. The pancreas contains the islets of Langerhans, which house beta cells that produce insulin. The beta cells monitor glucose levels in the blood and release finely measured amounts of insulin to counterbalance glucose peaks. Type I and II diabetes develop when more than 90 percent of these beta cells are damaged.
Type 1 diabetes is a disorder of glucose homeostasis that affects -10% of the 21 million diabetic individuals in the USA. Type 1 diabetes results from the virtually complete autoimmune destruction of pancreatic islet β cells, leaving individuals dependent upon the administration of insulin to maintain life. The pathogenesis of Type 1 diabetes involves a complex interplay between cells of the immune system and antigens present on the islet β cell. This interplay leads to the activation of ThI cells and macrophages, which subsequently release, among other factors, cytokines (ILl β, TNFα, and IFNγ) that induce nitric oxide production, which triggers both apoptotic and necrotic islet β cell death, β cell "replacement" strategies involving pancreas or islet transplantations have been variably successful in reversing type 1 diabetes, but require tissues from limiting sources (cadaveric donors) and are still subject to destruction by the underlying autoimmune process.
Separation or isolation of the islets from the connective matrix and remaining exocrine tissue is advantageous and beneficial for laboratory experimentation and transplantation purposes. Islet transplantation is a most promising and minimally physiologically invasive procedure for treatment of Type I diabetes mellitus. Transplanting islets rather than complete pancreatic tissue has the distinct advantages of ease of transplantation, and the elimination of the pancreatic exocrine function of the donor tissue involving secretion of digestive en2ymes. Liberating islets from pancreatic exocrine tissue is the initial and crucial step that influences islet transplantations. The important objective in islet isolation is to provide sufficient numbers of viable functional and potent islets for transplantation. The "Edmonton Protocol" transplants healthy islets into diabetic patients. Islet transplantation using the Edmonton Protocol is described in Shapiro, Ryan, and Lakey, Clinical Islet Transplantation-State of the Art, Transplantation Proceedings, 33, pp. 3502-3503 (2001); Ryan et al., Clinical Outcomes and Insulin Secretion After Islet Transplantation With the Edmonton Protocol, Diabetes, Vol. 50, April 2001, pp. 710-719; and Ryan elal., Continued Insulin Reserve Provides Long-Term Glycemic Control, Diabetes, Vol. 51, July 2002, pp. 2148- 2157. Once in the liver, the cells develop a blood supply and begin producing insulin. The Edmonton Protocol may include 7-10 steps depending on the method employed. The first step involves the delivery of a specific enzyme (liberase) to a donor pancreas, which digests the pancreas tissue, but does not digest the islets. Following the digestion step, there are several successive steps for separating the islets from other cells in the pancreas. The separated islets are transplanted into the main vessel of the liver, known as the portal vein. The liver is able to regenerate itself when damaged, building new blood vessels and supporting tissue. Therefore, when islets are transplanted into the liver, it is believed that new blood vessels form to support the islets. The insulin that the cells produce is absorbed into the blood stream through these surrounding vessels and distributed through the body to control glucose levels in the blood.
Altogether, the steps of the Edmonton Protocol create a vigorous process that compromises the viability of islets, which have a fragile, three-dimensional structure and require large amounts of oxygen for growth and viability. During the process, islets may be damaged or destroyed due to non-optimal conditions of oxygen delivery, affecting the yield of healthy islets that are retrieved from a given donor pancreas. Furthermore, islet transplantation is severely limited by donor availability; frequently, two pancreata are required to obtain insulin independence in just one patient.
Islet transplantation, together with steroid-free, nondiabetogenic immunosuppressive therapy, has been used to treat patients with Type 1 diabetes. However, such treatments can lead to increased risk of hyperlipidemia and hypertension, and long-term studies demonstrate that islet viability is impaired.
The Edmonton protocol of islet transplantation in humans has demonstrated remarkable short term success, with 80% of individuals achieving insulin independence at 1 year post- transplant. However, this rate decreases to only about 10-15% at 5 years. The reasons for this progressive loss of graft function is unclear, but likely includes islet loss as a result of ongoing cytokine-mediated inflammation. However, islet transplantation may continue to remain a viable therapeutic option for individuals with Type 1 diabetes, particularly as results show that even those that fail long-term insulin independence, they still retain other important benefits, such as decreased blood sugar lability, lower incidence of hypoglycemia, and decreased insulin dosage. The first days following islet transplantation are a particularly vulnerable period for the transplanted islets. Up to 60% of transplanted islets undergo non-autoantigen-specifϊc apoptosis in the first days following transplantation. This early destruction is the major underlying reason for requiring the use of two to three cadaveric donors to ensure sufficient islets survive to reverse diabetes. Local production of inflammatory cytokines IL- lβ, TNFα, and IFNγ have been implicated in the early apoptosis of transplanted islets. Thus, strategies that limit cytokine production in the early transplantation period may both limit the number of donors necessary to obtain sufficient islets, and also enhance islet survivability and prolong their ability to function in the long term. The present invention provides these strategies.
SUMMARY OF THE INVENTION
The present invention provides a method for preserving the functionality of harvested islet cells after isolation comprising administering eIF-5Al siRNA to the islet cells of an islet cell donor prior to islet isolation, wherein the eIF-5Al siRNA inhibits expression of eIF-5Al in the islet cells and thereby inhibits apoptosis in the islet cells and preserves the functionality of the harvested islet cells. In preferred embodiments the siRNA targets the following nucleotide sequences of eIF-5Al: 5'-AAAGGAATGACTTCCAGCTGA-S'; 5'- AAGATCGTCGAGATGTCTACT-3'; 5'-AAGGTCCATCTGGTTGGTATT-S'; or 5'- AAGCTGGACTCCTCCTACACA-3'. In certain embodiments the eIF-5Al siRNA comprises the nucleotide sequence 5'-AAAGGAAUGACUUCCAGCTGAdTdT-S'. The siRNA may be administered to the donor through any acceptable means. In certain embodiments, the siRNA is administered to the islet cell donor via intraperotoneal injection.
Another embodiment of the present invention provides a method for inhibiting islet cells from undergoing apoptosis during a donor harvesting process comprising administering eIF-5Al siRNA to an islet cell donor via intraperotoneal injection to islet cell donor prior to islet isolation, wherein the eIF-5Al siRNA inhibits expression of eIF-5Al in the islet cells and thereby inhibits apoptosis in the islet cells. In preferred embodiments the siRNA targets the following nucleotide sequences of elF- 5Al: 5'-AAAGGAATGACTTCCAGCTGA-S'; 5'-AAGATCGTCGAGATGTCTACT-S'; 5'- AAGGTCCATCTGGTTGGTATT-S'; or 5'-AAGCTGGACTCCTCCTACACA-S'. In certain embodiments the eIF-5Al siRNA comprises the nucleotide sequence 5'- AAAGGAAUGACUUCCAGCTGAdTdT-3'.
Any siRNA or antisense construct can be used, as long as such construct inhibits expression of eIF-5Al in the islet cells. Administration of siRNA may be by any suitable route. Exemplary administration methods include perfusion through the portal vein of the islet cell donor, hydrodynamic perfusion through the portal vein of the islet cell donor, and intraperotoneal administration.
The present invention also provides a composition for inhibiting apoptosis and preserving functionality of in islet cells, comprising eIF-5Al siRNA, wherein the siRNA inhibits expression of eIF-5Al and thereby inhibits apoptosis in the islet cells. Preferred eIF-5Al siRNAs are discussed above. The present invention also provides a method of inhibiting expression of a target protein in islet β cells comprising administering to a β cell donor prior to β cell harvesting an siRNA construct targeted to the mRNA encoding the target protein, wherein the siRNA inhibits expression of the target protein in the islet β cells.
The invention also provides a method of decreasing the production of nitric oxide (NO) via the inducible nitric oxide synthase (iNOS) pathway, by decreasing expression of eIF-FA.
The invention also provide the use of eIF5A siRNA to manufacture a medicament to decrease production of nitric oxide via the iNOS pathway.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 provides results of RT-PCR performed for β-actin, mAAT and eIF-5Al after perfusion through the portal vein with eIF-5A siRNA. This figure shows that eIF-5Al expression is measurable and was thus incorporated into islets.
Figure 2 shows slows retrograde portal vein perfusion. Bile duct (clear) and portal vein (red) ready for preparatory knot (dark suture). The needle enters below the knot (direction indicated by arrow), cross under the knot and releases siRNA into vessels that reach pancreas, spleen, intestine and a third of distal colon. Figure 3 shows that perfusion of eIF-5Al siRNA into islets causes a reduction of expression of eIF-5Al (shown is reduction in mRNA levels of eIF-5Al).
Figure 4 shows a reduction of apoptosis of islets cells having been treated with eIF5-5Al siRNA as compared to control and saline treated islets (here n=2 per group). Figure 5 shows a reduction of apoptosis of islets cells having been treated with eIF5-5Al siRNA as compared to control and saline treated islets (here n=3 per group).
Figure 6 provides the nucleotide sequence of human eIF-5Al aligned against eIF5-A2.
Figure 7 provides the amino acid sequence of human eIF-5Al aligned against eIF5-A2.
Figure 8 provides the nucleotide sequence of human eIF-5Al with exemplary antisense oligonucleotides.
Figure 9 provides the nucleotide sequence of human eIF-5Al with exemplary antisense oligonucleotides.
Figures 1OA and B provide the nucleotide sequence of human eIF-5Al with exemplary siRNAs. Figure 11 provides the nucleotide sequence of human eIF-5Al with exemplary siRNAs.
Figure 12 shows a schematic of experiments showing that siRNA eIF-5Al was able to knock down (decrease) expression in islet cells.
Figure 13A shows the pattern of insulin secretion for 50 islets and Figure 13B shows the pattern of [Ca2+]I from a single islet following 2.8 to 11 mM glucose stimulation. The phases are by 0, 1, 2. This figure shows a normally functioning islet responds to glucose stimulation with a biphasic pattern of insulin secretion, which closely resembles similar changes in islet intracellular calcium ([Ca2+];).
Figure 14 provides the results of calcium oscillation studies. These figures show that islet cells treated with eIF-5Al siRNA (and thus have decreased expression of eIF-5Al) showed a significantly stronger glucose response and overall oscillation. This indicates that these treated cells not only survive longer but also retain functionality.
Figure 15 shows different photographs of stained cytokine treated islet cells. The photographs shows that islet cells treated with eIF-5Al siRNA have more live and less dead cells than the control or vehicle treated (saline only) islet cells. Figures 16A and 16B show through four different imaging techniques that the siRNA gets into islets cells when administered to the islet cell donor via IP injection. Confocal images were used to generate 3D reconstructions of isolated islets. Figure 16A shows that islets isolated from mice administered 0.9% saline intraperitoneally once daily for three days show characteristic FITC autofluorescence but minimal fluorescence in the TRITC channel (labeled
Cy3 in figure). Figure 16B shows that islets isolated from C57BI/6 mice administered Cy3- labeled stabilized siRNA intraperitoneally once daily for three days show penetrance of the Cy3 label.
Figure 17 (A) shows the results of where βTC3 cells were transfected with two different siRNA constructs (constructs A and B) or controls and subjected to immunoblotting for the indicated proteins. Figure 17 (B) shows the results of where C57B1/6 mice were injected with the same siRNAs intraperitoneally once daily for three days, and islets were isolated and purified via collagenase digestion and differential gradient centrifugation. Islets were lysed in 2% SDS and subjected to immunoblotting. Figure 17 (C) shows the results where isolated islets were loaded with Fura2 for 30 minutes before imaging in 3mM D-glucose. Islets were stimulated with 1 ImM D-glucose at ~300 seconds, and the Fura2 ratio was continuously monitored by fluorescence microscopy. Figure 17(D) shows the results where glucose stimulated insulin secretion (GSIS) was performed using 50 islets from each treatment group.
Figures 18A-I show that knockdown of eIF5A improvise islet function and survival ex- vivo.
Figures 19A-D show that cytokine induced of iNOS protein production is absent in eIF5A-deficient cells.
DETAILED DESCRIPTION OF THE INVENTION
Eukaryotic initiation factor 5A (eIF5A) appears to be a crucial factor in the post- transcriptional regulation of stress-induced genes, and appears to promote cytokine-mediated apoptosis in mammalian cells. eIF5A is a small acidic protein that is very highly conserved throughout eukaryotes, and is the only protein known to contain the unique polyamine-derived amino acid hypusine. eIF5 A has been proposed to play roles in mRNA processing, trafficking, and translation. However, its role, if any, in the inflammatory cascade in pancreatic islets has not before been characterized. Pancreatic islets are highly vulnerable to glucose and lipid toxicity, oxidative stress, and inflammatory cytokines. To study the role of factors involved in mediating islet to response cellular stress, the present inventors developed a protocol to deplete islets in vivo of specific proteins using small interfering RNAs (siRNAs). In a study, the inventors sought to determine the role of eIF5A in islets by depleting them of eIF5A protein using RNA interference.
However, a major challenge in RNA interference studies in primary rodent islets is their relatively poor transfection efficiency. Whereas previous work has demonstrated the utility of viral-mediated delivery of short hairpin RNAs, this technique is time-consuming to generate the appropriate viruses and is subject to viral toxicity. The inventors discovered a novel protocol to deplete mouse islets of selected proteins in vivo by repeated intraperitoneal injections of stabilized small interfering RNAs (siRNAs). This protocol results in remarkable penetration of siRNAs within the islet. This technique can be used to successfully deplete islets of any desired/target protein such as the well-characterized pancreatic transcription factor Pdxl . Using this technique, the inventors have also successfully depleted islets of eIF5A protein, and demonstrated that eIF5A contributes to cytokine-mediated islet dysfunction by promoting translation of the mRNA encoding inducible nitric oxide synthase (iNOS). The data suggest a model whereby eIF5 A is necessary in pancreatic islets for stabilization and/or nuclear export of a subset of mRNAs (possibly those involved in cytokine-mediated stress responses), including iNOS mRNA.
The inventors employed in vivo siRNA injection to study the role of eukaryotic initiation factor 5A (eIF5A) in cytokine-mediated stress. eIF5A has been characterized in other systems as a crucial regulator in the translation of stress-induced genes. When siRNA against eIF5A was injected into mice, a 50-70% decrease in eIF5A protein levels in isolated islets was observed. Accompanying this fall, a 30-40% increase in glucose stimulated insulin secretion and Ca2+ mobilization as compared to control treatment was observed. When isolated islets were exposed to a cocktail of proinflammatory cytokines (ILl β, TNFα, IFNγ), the relative enhancement of islet function persisted in animals treated with si-eIF5A. Islet protection following eIF5A knockdown was not accompanied by alterations in the expression of genes whose products are involved in glucose responsiveness or insulin transcription (Slc2a2, Gck, Irsl, Nkx6-1, MafA, Pdxl, NeuroDl, and Setd7). See Figure 18C. Importantly, although the mRNA encoding inducible nitric oxide synthase (iNOS) was upregulated more than 40-fold in both control- and si-eIF5A- treated islets in response to cytokines (see figure 19A), iNOS protein levels were 3-5-fold lower in si-eIF5A-treated islets (see figure 19B). These data suggest that enhanced function in si- eIF5A-treated islets may be secondary to reduced nitric oxide production. This provides evidence that eIF5A plays an essential role in islet β cell response to stress signals via control of iNOS translation, and as such, eIF5A may serve as a viable target for therapeutic strategies aimed at preserving viability as well as the ability of the islet cells to function after harvesting and post implantation.
The inventors have further shown that blockage of eIF-5A function via inhibition of hypusination (via administration of GC-7 - an inhibitor of DHS) effectively reduces iNOS levels). See figure 19D.
It has been previously shown that siRNA incorporation into islets can be achieved by pancreatic perfusion via retrograde portal vein inoculation. See Bradley, et al., Transplantation Proceedings, 37, 233-236, 2005. Briefly, Cy-3 labeled Luciferase (Luc) siRNA GL2 duplex was used either packaged with Lipofectamine 2000 or unpackaged, and injected either through tail vein (in vivo, 50 μg per mouse) or directly into the pancreas by retrograde portal vein inoculation (in situ, 2 μg per mouse). Pancreata were procured and stored at 40C for 24 hours after in situ delivery, or 4 hours after in vivo delivery, and islets were isolated and cultured an extra 16 hours before examination. To visualize siRNA distribution, pancreata were stained for insulin and examined under a fluorescent microscope. Isolated islets were directly examined under a fluorescent microscope. Unpackaged siRNA reached islets to a similar extent as observed using liposomal-packaged siRNA, agreeing with reports of so-called "naked"-siRNA delivery in vivo. Lewis et al., Nat. Genet. 32: 107- 108, Epub 2002 JuI 2029, 2002 and
McCaffrey AP, et al., Nature 418:38-39, 2002). Figure 1 shows that perfusion to the islet cells provides a suitable delivery mechanism to the islet cells.
Accordingly, the present invention provides a method for inhibiting expression of a target protein in islet cells comprising administering siRNA targeted to the mRNA encoding the target protein to the islet cells, wherein the siRNA inhibits expression of the target protein in the islet cells. Administration may be through any means, preferably trough intraperitoneal administration to the islet β cell donor before the islet β cell isolation.
In one experiment, the inventors have shown that three daily intraperitoneal injections of stabilized, Cy3 -labeled double-stranded RNAs into C57BL/6 mice revealed remarkable penetration into isolated islets, as determined by confocal microscopy. To test this technique for protein knockdown, stabilized siRNAs targeted against Pdxl were used. Two distinct siRNAs against Pdxl message resulted in 50% and 90% knockdown of Pdxl protein in islets (as assessed by immunoblot analysis). See figures 17 A-D.
The present invention provides a method for inhibiting expression of eIF-5Al in islet cells comprising administering eIF-5Al siRNA to the islet cells, wherein the eIF-5Al siRNA inhibits expression of eIF-5Al in the islet cells. Figures 16A and 16B show that administration to the islet cell donor via IP injection also provides a suitable delivery mechanism to the islet cells. Figure 16B shows that three daily intraperitoneal injections of stabilized, Cy3-labeled double-stranded RNAs into C57BL/6 mice revealed remarkable penetration into isolated islets, as determined by confocal microscopy. Figures 12B and 18B show that the eIF-5Al siRNA treated islet cells do indeed express less eIF-5Al siRNA.
By inhibiting eIF-5Al expression, apoptosis is also inhibited. Figures 4 and 5 show that treating islets cells with eIF-5Al siRNA prior to isolation provided a mechanism to inhibit these cells from apoptosis (as demonstrated by a reduction of the number of cells in the sub-Gl phase). Accordingly, the present invention also provides a method for inhibiting apoptosis in harvested islet cells comprising administering eIF-5 Al siRNA to the islet β cell donor, wherein the elF- 5Al siRNA inhibits expression of eIF-5Al in the islet cells and wherein the inhibition of elF- 5Al expression inhibits apoptosis. Preferably the islets cells are treated with eIF-5Al siRNA cells prior to the harvesting process.
The present invention further provides a method for preserving the functionality of harvested islet cells after isolation comprising administering eIF-5Al siRNA to the islet cells of an islet cell donor prior to islet isolation, which results in a inhibition or reduction of eIF-5Al expression in the islet cells, which in turn inhibits apoptosis in the islet cells, as well as preserves their functionality. Preserving the functionality of the islet cells means that not only do the islet cells survive the harvesting process, but also maintain their function (the functionality of the cells remains longer and/or they maintain a better glucose response than non eIF-5Al siRNA treated islet cells)(i.e. release insulin in response to glucose) after the harvesting process. In one experiment, to investigate the consequence of eIF-5Al knockdown on islet function and overall health, C57BL/6 male mice were given intraperitoneal (IP) injections of either: vehicle (0.9% saline) [Group I], control siRNA (16.6mg/kg) [Group H], or eIF-5Al siRNA (16.6mg /kg) [Group HI]. Injections of the assigned treatment were administered each day for three days (days -3, -2, -V) prior to islet harvest (day 0). Isolated islets were purified by collagenase treatment and allowed to rest for 16-18 hours before testing commenced. After establishing a baseline of function on Day 1 post-harvest, half of the islets were exposed to a cytokine cocktail (5ng/mL ILl β, lOng/mL TNF-α, lOOng/mL INF-γ) to mimic conditions pancreatic islets encounter in diabetes Type 1. A schematic of the experimental design and allocation of islets is depicted in Figure 12 A.
Protein lysates from each group of islets were analyzed by Western Blot for protein knockdown. Figure 12B illustrates the relative knockdown of eIF-5Al in Group III treated mouse islets, which is quantified in Figure 12C. These data illustrate the efficacy of IP injection to produce knockdown expression of eIF-5Al in islets. Evidence of preserved functionality of the harvested islets is demonstrated by calcium oscillation assays. The inventors have determined that intracellular calcium is an indicator of stimulation and stress. For both humans and rodents, a normally functioning islet responds to glucose stimulation with a biphasic pattern of insulin secretion, as shown in Figure 13 A. As a first approximation, this biphasic secretion pattern closely resembles similar changes in islet intracellular calcium ([Ca2+]O, which also consists of a first phase peak that drops into a second phase plateau (Figure 13B). Measurements of [Ca2+]; reported as the ratio of emitted light from 340 and 380 nm stimulation, however, provide greater sensitivity to detect additional features of islet function. The [Ca2+]; response to glucose consists of three phases (see figure 13) that roughly reflects the processes described by the "Consensus Model" of beta-cell stimulus- secretion coupling. An initial dip in [Ca2+]i occurs following glucose transport into the beta-cell as the endoplasmic reticulum mobilizes and sequesters [Ca2+Jj (phase 0). Glucose is then metabolized through glycolysis and further metabolized in the tri-carboxylic acid (TCA) cycle, which results in ATP production. When a sufficient increase in ATP/ADP occurs to close KATP- ion-channels, a sharp rise in [Ca2+]; occurs due to the opening of L-type calcium channels related to first phase insulin secretion. After this initial first phase peak, [Ca2+]i drops to a plateau that is often punctuated by [Ca2+]; oscillations. The plateau height is directly related to glucose concentration and the rate of second phase insulin secretion. Overall, this imaging-based [Ca2+]; measurement is thus a good first approximation of dynamic insulin secretion at the level of the individual islet. Many factors can negatively impact the normal [Ca2+]; response to glucose stimulation.
Disruptions in glucose metabolism, mitochondria and ATP production, endoplasmic reticulum (ER), ion channel function, and myriad other problems can affect the dynamics of calcium handling related to glucose stimulation. By frequently monitoring changes in [Ca2+Ji, defects in these aspects of activity are more likely to be observed than by standard measures of static insulin secretion. For example, increased basal [Ca2+J1 in low glucose and the loss of the phase 0 dip in [Ca2+J; during glucose stimulation can be indicative of ER-stress or possibly ion-channel dysfunction. Such defects can not be detected easily by measuring static or kinetic insulin secretion, but the imaging techniques described herein can be utilized to address these questions.
In both the presence and absence of cytokines, in calcium oscillations and glucose stimulated insulin secretions studies, the eIF-5Al siRNA (16.6 mg/kg) treated islets [Group III (eIF-5Al knockdown islets)] showed a greater response to glucose and an extended maintenance of functionality (implying extended survival). See Figure 14. In accord, these islets are also expected to show a higher production of insulin (data pending). Preliminary data from images taken after Live/Dead staining qualitatively support the notion of eIF-5Al knockdown extending the life of islet cells (Figure 15). By comparing the number of red islets, the result of EtHD-I intercalation into the nuclei of dead islet cells, to the number of green islets that are actively cleaving calcein-AM into the fluorescent calcein, a qualitative image of islet viability is ascertained (Figure 15).
The present invention further provides a method of inhibiting apoptosis in cells involved in inflammtory diseases in which the disease is associated with increased production of nitric oxide. For example, inflammatory diseases such as rheumatoid arthritis (RA) are associated with increased production of nitric oxide (NO), due to activation of the inducible nitric oxide synthase (iNOS) pathway. Studies in animal models have suggested that NO plays a causal role in the pathogenesis of joint inflammation and tissue damage, since the severity of arthritis can be reduced by the administration of NOS inhibitors. Several cell types present within the joint, including synovial fibroblasts, endothelial cells and chondrocytes, can be induced by pro- inflammatory cytokines to produce NO in vitro. Moreover, localization studies have shown that there is up-regulation of iNOS expression in synovial lining cells, chondrocytes and blood vessels in joint tissues obtained from patients with RA. The localization of iNOS expression to the synovial lining layer and cartilage is of interest in the light of other studies that have shown that apoptosis is increased in RA, particularly in the synovial lining layer and cartilage. The mechanisms responsible for apoptosis in the rheumatoid joint remain unclear, although previous workers have shown that expression of Fas antigen is increased in RA synoviocytes and that Fas antibody can stimulate the apoptotic death of synoviocytes in vitro. Others have investigated the hypothesis that activation of the iNOS pathway is also involved in stimulating apoptosis in the rheumatoid joint, since high levels of NO are known to stimulate apoptosis in many cell types in vitro and have concluded that NO acts as a mediator of apoptosis in RA. See R. J. van't Hof et al., Rheumatology 200, 39:1004-1008.
As such, the present invention provides a method of inhibiting apoptosis in cells associated with RA (such as synovial and cartilage cells) by inhibiting expression of eIF5A and hence inhibits or decreases the production of nitric oxide via the iNOS pathway to inhibit apoptosis in the cells.
The present invention further provides a method of inhibiting NO production in a cell or host by administration to the cell or host an siRNA directed against eIF-5A. The siRNA decreases expression of eIF-5A and in turn decreases the production of NO by inhibiting the activation of the iNOS pathway. The invention also provide the use of eIF5 A siRNA to manufacture a medicament to decrease production of nitric oxide via the iNOS pathway.
Any eIF-5Al siRNA that inhibits expression of eIF-5Al may be used. The term "inhibits" also means reduce as compared to levels that would occur in control cells, i.e. those cells not having been treated with eIF-5Al siRNA. One exemplary and especially preferred elF- 5Al siRNA comprises the sequence: 5'-AAAGGAAUGACUUCCAGCTGAdTdT-S'. Co- pending application 11/293,391, which was filed on November 28, 2005 (which is herein incorporated by reference in its entirety) provides additional exemplary eIF-5Al siRNAs and other antisense constructs that have been used to inhibit expression of eIF-5Al in other cell types and were also shown to inhibit apoptosis. One skilled in the art could design other eIF-5Al siRNAs given the eIF-5Al sequence and can easily test for the siRNAs ability to inhibit expression without undue experimentation. Figures 6-11 provide sequences of eIF-5Al, exemplary eIF-5Al siRNAs and antisense constructs. In another embodiment of the invention, antisense constructs of eIF-5Al may be used to inhibit expression of eIF-5Al and thus inhibit apoptosis of the islet cells, as well as maintain or preserve their functionality. In preferred embodiments the eIF-5Al siRNA targets the following nucleotide sequence ofeIF-5Al (see figure 10): 5'-AAAGGAATGACTTCCAGCTGA-S'; 5'-AAGATCGTCGAGATGTCTACT-S'; 5'-AAGGTCCATCTGGTTGGTATT-S'; and 5'- AAGCTGGACTCCTCCTACACA-3'. In especially preferred embodiments, the siRNA targets the following sequence of eIF-5Al: 5'-AAAGGAATGACTTCCAGCTGA-S'.
The present invention also provides a method for inhibiting islet cells from undergoing apoptosis during a donor harvesting process. As discussed above, many islets cells undergo apoptosis when they are harvested. The present inventors have shown that providing eIF-5Al siRNA to the islet cells prior to harvesting, offers a protective benefit against apoptosis. The eIF-5Al siRNA is administered to the islet cells of an islet cell donor prior to islet isolation. The donor (and hence islet cells) may be any animal, including human islet cells. Any method of administration may be used. For example, the siRNA may be administered via perfusion through the portal vein of the islet cell donor or via hydrodynamic perfusion through the portal vein of the islet cell donor. See example 1. Another form of administration includes intraperotoneal administration. See example 2 and examples 3-5.
Perfusion through portal vein is similar to canulation of the bile duct, but the needle points the opposite way. The portal vein is exposed by retraction of liver and shifting of visceral organs to the mouse's left. A preparative knot is made around it and includes the bile duct. After puncturing the vessel a blunted needle is advanced toward the pancreas and the knot is tightened around it. In a mouse model, 1 ml saline or siRNA (5 μg) is released slowly, the needle is removed and the knot is closed behind the needle to prevent fluid escape. At this point the mouse is turned around and the bile duct accessed for pancreas digestion. The pancreas may be held longer with siRNA. Alternatively, it can be removed but kept cold with collagenase longer. Regular islet isolation methods are followed and the islets (50) may be incubated in for 16 hours.
The present invention also provides a composition for inhibiting apoptosis in islet cells, comprising eIF-5Al siRNA, wherein the siRNA inhibits expression of eIF-5Al and thereby inhibits apoptosis and maintains the functionality of the islet cells. The composition may comprise other or additional eIF-5Al siRNAs as discussed above. A preferred siRNA comprises the nucleotide sequence 5'-AAAGGAAUGACUUCCAGCTGAdTdT-S' EXAMPLES
Example 1 : Portal vein perfusion
Mouse islets express elFSA. Total RNA was extracted from isolated mouse islets and RT-PCR was performed for (β-actin and for eIF-5Al (fig. 1). Resting non-stimulated islets exhibited positive levels of eIF-5Al-mRNA. elFSAl-mRNA levels diminished after elF5Al-siRNA delivery: portal vein slow perfusion. Mice were introduced 1 ml of siRNA (CT (control) sequence or eIF-5Al, 5 μg) or saline, n = 2 per group, by slow retrograde portal vein perfusion (fig. 2). Pancreata were digested by collagenase irrigation of pancreatic duct and islets were isolated as described by
Lewis et al., Proc. Natl. Acad. Set USA, 102:12153-12158 Epub 12005 Aug. 12110, 2005. Islets (50 per mouse) were incubated for 16 hours. Total RNA was then extracted and RT-PCR was performed for β-actin and for eIF-5Al (fig. 3). Ratio of mRNA for eIF~5Al/β-actin was 5.24 (CT-siRNA) and 3.01 (eIF-5Al-siRNA). Figure 3 shows that mRNA levels of eIF-5Al were reduced in those cells treated with siRNA. This experiment was repeated with n = 3 mice and islets were incubated for RNA extraction in triplicates; results were consistent with initial observation. eIF-5Al-mRNA levels diminished and islet apoptosis rate reduced after elF5A-siRNA delivery: portal vein hydrodynamic perfusion. Mice were introduced 1 ml of siRNA (CT or elF- 5Al, 5 μg) or saline, n = 2 per group, by hydrodynamic retrograde portal vein perfusion, which was completed within 5 seconds. Pancreata were digested by collagenase irrigation of pancreatic duct and islets were isolated. Islets were incubated for 16 hours and then divided: one group was stained with propidium iodide for evaluation of apoptosis (50 islets per mouse) and the other group was processed for RT-PCR (25 islets per mouse). Levels of mRNA for eIF-5Al/β-actin were again higher in CT-siRNA group than in eIF-5 Al -siRNA group. Apoptosis rate was reduced by 28.1% (fig.4). This experiment was repeated with n = 3, apoptosis rate again diminished (fig. 5).
Islets perfusion with Uotinylated-siRNA. Biotinylated-siRNA (50 μg) was perfused into islets as described above (slow perfusion, n = 1). Pancreas was fixed in formalin for staining. siRNA. siRNA molecules were synthesized by Dhaimacon, Lafayette, CO. The sequence of the eIF-5Al and control siRNA were: 5' CGGAAUGACUUCCAGCUGAdTdT 31 and 5' AGUCGACCUUCAGUAAGGCdTdT 3', respectively.
RT-PCR. Total RNA was extracted from cells using Qiagen RNeasy kit. eIF-5Al Primers: Forward 5'-GAC AGT GGG GAG GTA CGA GA-3'; Reverse 5'-GGG GTG AGG AAA ACC AAA AT-31.
Propidium iodide (PI) apoptosis stain. Single cell suspension of islets was achieved by gentle trypsinization. Cells were washed with PBS and added saponin-PI mixture containing 0.3 % Saponin, EDTA 1 mM, Rnase, 1 % Azide, 1 % FCS and 50 μg/ml PI in PBS. Cells were thoroughly vortexed and incubated at 40C in the dark for 6 hours before analyzed for sub-GI population by FACS.
Example 2: intraperotoneal injection
Antibodies and siRNA: Mouse monoclonal antibody against eIF-5Al were generated. Anti-actin monoclonal antibody (clone C4, #69100) was purchased from MP Biomedicals. In Western blots, near infrared fluorophore labeled secondary antibodies from Li-Cor were used (IRDye 800 and IRDye 700). The siRNA specific for eIF-5Al was directed to the following sequence of eIF-5Al : 5'-AAAGGAATGACTTCCAGCTGA-S'. While control siRNA was the transcribed sequence, 5'-AAAGTCGACCTTCAGTAAGGA-S', and was previously shown to not induce knockdown of any known protein. See co-pending applications U.S. 11/725,470 and PCT/US07/64424, herein incorporated by reference in their entireties.
IP (intraperotoneal) Injection of mice with siRNA: Twelve, 8- to 10-week old C573L/6 male mice were purchased from Charles River. Male mice were chosen to avoid the metabolic variation that accompanies the female estrous cycle. Mice were randomly assigned one of three treatment groups: Group I - vehicle treatment (0.9% saline), Group II - control siRNA treatment, Group III - eIF-5Al siRNA treatment. Each mouse received the select treatment via IP injection with 16.6mg/kg or similar volume (in the case of Group I). Mice were injected each day (approximately 11 :00 a.m.) for three days (Days -3, -2, -1) prior to islet harvest (Day 0). Islet Isolations and Cytokine Treatment: Islet isolations from C57BL/6 mice were carried out using standard isolation techniques and a protocol approved by the Institutional
Animal Care and Use Committee (IACUC). Pancreata were stored in HBSS supplemented with 4.2mM sodium bicarbonate and 1% BSA (Invitrogen) until collagenase digestion. Purification was performed by differential centrifugation. Whole islets were maintained in phenolred-free DMEM solution supplemented with 10% FBS and 5% penicillinn/streptomycin. Cultures were incubated at 370C + 5% CO2 in 5.5mM glucose. Purified islets were allowed to recover for 16- 18 hours before metabolic testing (calcium oscillations).
Cytokine treatment was applied to half of the islets at the end of metabolic testing on Day 1 using a prescribed cocktail of physiologically relevant cytokines (5ng/mL IL lβ, lOng/mL TNT-α, 100ng/mL INF-γ. Cytokine treated islets were allowed to rest 24 hours before further testing and protein analysis commenced (Day 2 and Day 3). Immunoblot Assays: 10 μg of islet cell extract (prepared in Laemmli buffer containing
20OmM DTT and Benzonase) were resolved by electrophoresis on a 4-20% SDS-polyacrylamide gel (Invitrogen) followed by immunoblot analysis using anti-eIF-5Al mouse monoclonal primary antibody (at a 1 : 15000 dilution). Immunoblots were imaged and quantitated with the near infra red technology of the Odyssey system (Li-Cor Biosciences). Calcium Oscillation Assays: Pancreatic islets were handpicked from collagenase-digested
8- to 10-wk-old C57BL6 mouse pancreas (as described above). Islets were treated for 20 minutes in Fura-2 low glucose (3mM) solution then placed in a temperature-controlled flow chamber. Low glucose solution (without Fura) was flowed over the islet to establish a basal calcium measurement (5 minutes). Measurements of calcium were obtained using IP Lab 4.0 software (BD Biosciences) and are reported as the ratio of emitted light from 340 to 380 nm.
After basal calcium levels were established, a high glucose (1 ImM) solution was flowed through the chamber (15 minutes) to induce the cellular calcium response of the islets. The calcium response was monitored in 5-second intervals over the 20-minute period of observation. Islets are disposed of in a 5% bleach solution upon conclusion of testing.
Example 3 : Application of siRNA against Pdx-1 in vitro and in vivo βTC3 cells were transfected with two different Pdx-1 siRNA constructs (constructs A and B) or controls and subjected to immunoblotting for the proteins Pdx-1, GAPDH and β-actin. See See Figure 17A. C57B1/6 mice were injected with the same siRNAs intraperitoneally once daily for three days, and islets were isolated and purified via collagenase digestion and differential gradient centrifugation. Islets were lysed in 2% SDS and subjected to immunoblotting. See Figure 17B. Isolated islets were loaded with Fura2 for 30 minutes before imaging in 3mM D- glucose. Islets were stimulated with 1 ImM D-glucose at ~300 seconds, and the Fura2 ratio was continuously monitored by fluorescence microscopy. See Figure 17C. Glucose stimulated insulin secretion (GSIS) was performed using 50 islets from each treatment group. See Figure 17D.
Example 4: Knockdown of eIF-5A improves islet function and survival ex vivo
This example shows that in vivo administration of siRNA against eIF5A improves islet function ex vivo. Figure 18A provides the schematic representation of experimental design. Islets were isolated post-treatment and extracts were subjected to immunoblot analysis. See figure 18B. Isolated islets were subjected to RT-PCR analysis of genes essential to glucose sensing and insulin transcription; although gene transcription was inhibited by cytokine treatment (4 h incubation with IFNg, ILIb, TNFa), no differences between groups were observed. See Figure 18C. si-eIF5A-treated islets 24h (see figure 18D) and 48h (see figure 18F) after isolation show improved Glucose Stimulated Calcium responses (GSCa) compared to controls at 24 h (see figure 18D) and 48 h (see figure 18F) after isolatin. si-eIF5A-treated islets demonstrate enhanced GSCa in the presence of cytokines 24h (see figure 18E) and 48h (see figure 18G) after isolation. Islets isolated from eIF5A treated mice showed improved glucose stimulated insulin secretion ("GSIS") compared to controls in both the absence (see figure 18H) and presence (see figure 18H) of 4h cytokine incubation, as assessed by GSIS.
Example 5: Knockdown of eIF-5A abrogates cytokine-induced INOS production
Cytokine induced of iNOS protein production is absent in eIF5A-deficient cells. Islets treated with cytokines were subjected to RT-PCR for iNOS mRNA; all treatment groups showed dramatic upregulation of iNOS mRNA upon cytokine exposure. See figure 19A. Cytokine- treated islets were subjected to immunoblot analysis. Notably, iNOS protein was induced upon cytokine exposure in control islets, but not si-eIF5A-treated islets. See Figure 19B. INS-I (832/13) β cell were treated with cytokine cocktail for 4 h and subjected to immunoblotting; cells show similar increases in iNOS protein. See figure 19C. INS-I cells were treated with varying concentrations of GC-7, an inhibitor of deoxyhypusine synthase, overnight, then incubated for 4h in the presence of cytokines. INS-I cells show an inverse correlation of iNOS production and GC-7 concentration, suggesting that inhibition of active eIF5A production prevents iNOS translation. See figure 19D.
References
1. FarreII AJ, Blake DR, Palmer RMJ, Moncada S. Increased concentrations of nitrite in synovial fluid and serum samples suggest increased nitric oxide synthesis in rheumatic diseases. Ann Rheum Disl992;51 : 1219-22.
2. Grabowski PS, England AJ, Dykhuizen R et al. Elevated nitric oxide production in rheumatoid arthritis: detection using the fasting urinary nitrite/creatinine ratio. Eur J Pharmacoll996;l 10:701-6.
3. Hilliquin P, Borderie D, Hernvann A, Menkes CJ, Ekindjian OG. Nitric oxide as S- nitrosoproteins in rheumatoid arthritis. Arthritis Rheuml997;40:1512-7.
4. McCartney-Francis N, Allen JB, Mizel DE et al. Suppression of arthritis by an inhibitor of nitric oxide synthase. J Exp Medl993;178:749~54. 5. Stefanovic-Racic M, Meyers K, Meschter C, Coffey JW, Hoffman RA, Evans CH. N- monomethyl arginine, an inhibitor of nitric oxide synthase, suppresses the development of adjuvant arthritis in rats. Arthritis Rheuml994;37:1062-9.
6. Ialenti A, Moncada S, Di Rosa M. Modulation of adjuvant arthritis by endogenous nitric oxide. Br J Pharmacol 1993 ;110:701-6. 7. Palmer RM5 Hickery MS, Charles IG, Moncada S, Bayliss MT. Induction of nitric oxide synthase in human chondrocytes. Biochem Biophys Res Communl993;193:398-405.
8. Stadler J, Stefanovic-Racic M, Billiar TR et al. Articular chondrocytes synthesize nitric oxide in response to cytokines and lipopolysaccharide. J Immunoll991;147:3915-20.
9. Grabowski PS, Macpherson H, Ralston SH. Nitric oxide production in cells derived from the human joint. Br J Rheuml996;35:207-12.
10. Ralston SH9 Todd D, Helfrich M, Benjamin N, Grabowski PS. Human osteoblast-like cells produce nitric oxide and express inducible nitric oxide synthase. Endocrinology 1994; 135:330-6.
11. Stefanovic-Racic M, Stadler J, Georgescu HI, Evans CH. Nitric oxide synthesis and its regulation by rabbit synoviocytes. J Rheumatoll994;21:1892-8.
12. Sakurai H, Kohsaka H, Liu M et al. Nitric oxide production and inducible nitric oxide synthase expression in inflammatory arthritis. J Clin Investl995;96:2357-63. 13. Grabowski PS, Wright PK, van 't Hof RJ, Helfrich MH, Oshima H, Ralston SH. Iraniunolocalisation of inducible nitric oxide synthase in the synovium and cartilage in rheumatoid arthritis and osteoarthritis. Br J Rheumatoll997;36:651-5.
14. Firestein GS, Yeo M, Zvaifler NJ. Apoptosis in rheumatoid arthritis synovium. J Clin Investl995;96:1631-8.
15. Nakajima T, Aono H, Hasunuma T et a Apoptosis and functional Fas antigen in rheumatoid arthritis synoviocytes. Arthritis Rheuml995;38:485-91.
16. Sioud M, Mellbye O, Forre O. Analysis of the NF-kappa B p65 subunit, Fas antigen, Fas ligand and Bcl-2-related proteins in the synovium of RA and polyarticular JRA. Clin Exp Rheumatoll998; 16: 125-34.
17. Brune B, Gotz C, Messmer UK, Sandau K, Hirvonen MR, Lapetina EG. Superoxide formation and macrophage resistance to nitric oxide-mediated apoptosis. J Biol Cheml997;272:7253-8.
18. Albina JE, Cui S, Mateo RB, Reichner JS. Nitric oxide-mediated apoptosis in murine peritoneal macrophages. J Immunoll993;150:5080-5.
19. Blanco FJ, Ochs RL, Schwarz H, Lotz M. Chondrocyte apoptosis induced by nitric oxide. Am J Patholl995;146:75-85.
20. Turpaev KT, Amchenkova AM, Narovlyansky AN. Two pathways of the nitric oxide- induced cytotoxic action. Biochem MoI Biol Intl997;41:1025-33.

Claims

1. A method for preserving the functionality of harvested islet cells after isolation comprising administering eIF-5Al siRNA to the islet cells of an islet cell donor prior to islet isolation, wherein the eIF-5Al siRNA inhibits expression of eIF-5Al in the islet cells and thereby inhibits apoptosis in the islet cells and preserves the functionality of the harvested islet cells.
2. The method of claim 1 wherein the eIF-5Al siRNA targets the following nucleotide sequence of eIF-5Al: 5'-AAAGGAATGACTTCCAGCTGA-S'; 5'-AAGATCGTCGAGATGTCTACT- 3'; 5'-AAGGTCCATCTGGTTGGTATT-S'; or 5'-AAGCTGGACTCCTCCTACACA-S'.
3. The method of claim 1 wherein the eIF-5Al siRNA comprises the nucleotide sequence 5'- A AAGGAAUGACUUCCAGCTGAdTdT-3 ' .
4. The method of claim 1 wherein the siRNA is administered via intraperotoneal injection to the islet cell donor.
5. A method for inhibiting islet cells from undergoing apoptosis during a donor harvesting process comprising administering eIF-5Al siRNA to an islet cell donor via intraperotoneal injection to islet cell donor prior to islet isolation, wherein the eIF-5Al siRNA inhibits expression of eIF-5 Al in the islet cells and thereby inhibits apoptosis in the islet cells.
6. The method of claim 5 wherein the eIF-5Al siRNA targets the following nucleotide sequence of eIF-5Al: 5'-AAAGGAATGACTTCCAGCTGA-S'; 5 '-AAGATCGTCGAGATGTCTACT- 3'; 5'-AAGGTCCATCTGGTTGGTATT-S'; or 5'-AAGCTGGACTCCTCCTACACA-S'.
7. The method of claim 5 wherein the eIF-5Al siRNA comprises the nucleotide sequence 5'- AAAGGAAUGACUUCCAGCTGAdTdT-3 '.
PCT/US2008/073642 2007-08-20 2008-08-20 Use of eif-5a1 sirna to protect islets cells from apoptosis and to preserve their functionality WO2009026317A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
AU2008288988A AU2008288988A1 (en) 2007-08-20 2008-08-20 Use of eIF-5A1 siRNA to protect islets cells from apoptosis and to preserve their functionality
JP2010521983A JP2010536379A (en) 2007-08-20 2008-08-20 Use of EIF-5A1 siRNA to protect islet cells from apoptosis and retain their function
EP08798216A EP2195429A2 (en) 2007-08-20 2008-08-20 Use of eif-5a1 sirna to protect islets cells from apoptosis and to preserve their functionality
CN2008801121650A CN102124108A (en) 2007-08-20 2008-08-20 Use of EIF-5A1 SIRNA to protect islets cells from apoptosis and to preserve their functionality
CA2700463A CA2700463A1 (en) 2007-08-20 2008-08-20 Use of eif-5a1 sirna to protect islets cells from apoptosis and to preserve their functionality
IL204072A IL204072A0 (en) 2007-08-20 2010-02-21 USE OF EIF - 5A1 siRNA TO PROTECT ISLETS CELLS FROM APOPTOSIS AND TO PRESERVE THEIR FUNCTIONALITY

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US95686707P 2007-08-20 2007-08-20
US60/956,867 2007-08-20
US4307408P 2008-04-07 2008-04-07
US61/043,074 2008-04-07

Publications (2)

Publication Number Publication Date
WO2009026317A2 true WO2009026317A2 (en) 2009-02-26
WO2009026317A3 WO2009026317A3 (en) 2009-07-16

Family

ID=40378961

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/073642 WO2009026317A2 (en) 2007-08-20 2008-08-20 Use of eif-5a1 sirna to protect islets cells from apoptosis and to preserve their functionality

Country Status (9)

Country Link
US (1) US20090093434A1 (en)
EP (1) EP2195429A2 (en)
JP (1) JP2010536379A (en)
KR (1) KR20100046266A (en)
CN (1) CN102124108A (en)
AU (1) AU2008288988A1 (en)
CA (1) CA2700463A1 (en)
IL (1) IL204072A0 (en)
WO (1) WO2009026317A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102782139A (en) * 2009-10-30 2012-11-14 格兰达利斯有限公司 Novel therapeutic RNA interference technology targeted to the PDX-1 oncogene in PDX-1 expressing neuroendocrine tumors
WO2015178490A1 (en) * 2014-05-23 2015-11-26 国立大学法人京都大学 Graft material and method for preparing same
CN105543259A (en) * 2009-10-30 2016-05-04 斯特莱科生物有限公司 Novel therapeutic RNA interference technology targeted to the PDX-1 oncogene in PDX-1 expressing neuroendocrine tumors
WO2021155025A1 (en) * 2020-01-28 2021-08-05 The Board Of Trustees Of The Leland Stanford Junior University Preventing or treating pancreatic dysfunction or diabetes by upregulating human cathelicidin

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005007853A2 (en) * 2003-06-06 2005-01-27 Senesco Technologies, Inc. Inhibition of apoptosis-specific elf-5a (“eif-5a1”) with antisense oligonucleotides and sirnas as anti-inflammatory therapeutics
WO2007109677A2 (en) * 2006-03-20 2007-09-27 Senesco Technologies, Inc. A novel method of protecting islet cells from apoptosis during the donor harvesting process

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7968523B2 (en) * 2001-07-23 2011-06-28 Senesco Technologies, Inc. Method for inducing apoptosis using apoptosis-specific EIF5-A

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005007853A2 (en) * 2003-06-06 2005-01-27 Senesco Technologies, Inc. Inhibition of apoptosis-specific elf-5a (“eif-5a1”) with antisense oligonucleotides and sirnas as anti-inflammatory therapeutics
WO2007109677A2 (en) * 2006-03-20 2007-09-27 Senesco Technologies, Inc. A novel method of protecting islet cells from apoptosis during the donor harvesting process

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: "senesco's new gene technology increases the survivability of pancreatic islet cells" DIABETES CURE BLOG, [Online] 23 March 2006 (2006-03-23), XP002524437 Retrieved from the Internet: URL:http://diabetes.taragana.net/senescos-new-gene-technology-increases-the-survivability-of-pancreatic-islet-cells/> [retrieved on 2009-04-21] *
ANONYMOUS: ANNUAL REPORT 2006 OF SENESCO TECHNOLOGIES, INC., [Online] 2006, pages 1-81, XP002524436 Retrieved from the Internet: URL:http://www.senesco.com/PDF/2006report.pdf> [retrieved on 2009-04-21] *
BRADLEY S P ET AL: "Successful incorporation of short-interfering RNA into islet cells by in situ perfusion" TRANSPLANTATION PROCEEDINGS, vol. 37, no. 1, 1 January 2005 (2005-01-01), pages 233-236, XP004819911 ISSN: 0041-1345 cited in the application *
CARAGLIA M ET AL: "The role of eukaryotic initiation factor 5A in the control of cell proliferation and apoptosis" AMINO ACIDS, vol. 20, no. 2, 1 January 2001 (2001-01-01), pages 91-104, XP002291822 ISSN: 0939-4451 *
LEWIS E C ET AL: "alpha1-Antitrypsin Monotherapy Prolongs Islet Allograft Survival in Mice" PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, vol. 102, no. 34, 23 August 2005 (2005-08-23), pages 12153-12158, XP003011675 ISSN: 0027-8424 cited in the application *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102782139A (en) * 2009-10-30 2012-11-14 格兰达利斯有限公司 Novel therapeutic RNA interference technology targeted to the PDX-1 oncogene in PDX-1 expressing neuroendocrine tumors
US9133459B2 (en) 2009-10-30 2015-09-15 Strike Bio, Inc. Therapeutic RNA interference technology targeted to the PDX-1 oncogene in PDX-1 expressing neuroendocrine tumors
CN102782139B (en) * 2009-10-30 2015-11-25 格兰达利斯有限公司 The new therapeutic RNA perturbation technique of target PDX-1 oncogene in the neuroendocrine tumor of expressing PDX-1
CN105543259A (en) * 2009-10-30 2016-05-04 斯特莱科生物有限公司 Novel therapeutic RNA interference technology targeted to the PDX-1 oncogene in PDX-1 expressing neuroendocrine tumors
WO2015178490A1 (en) * 2014-05-23 2015-11-26 国立大学法人京都大学 Graft material and method for preparing same
JPWO2015178490A1 (en) * 2014-05-23 2017-04-20 国立大学法人京都大学 Implant material and preparation method thereof
US11045425B2 (en) 2014-05-23 2021-06-29 Kyoto University Method of encapsulating pancreatic islet cells introduced to siRNA against PHLDA3
WO2021155025A1 (en) * 2020-01-28 2021-08-05 The Board Of Trustees Of The Leland Stanford Junior University Preventing or treating pancreatic dysfunction or diabetes by upregulating human cathelicidin

Also Published As

Publication number Publication date
US20090093434A1 (en) 2009-04-09
AU2008288988A1 (en) 2009-02-26
JP2010536379A (en) 2010-12-02
CA2700463A1 (en) 2009-02-26
WO2009026317A3 (en) 2009-07-16
CN102124108A (en) 2011-07-13
IL204072A0 (en) 2011-07-31
KR20100046266A (en) 2010-05-06
EP2195429A2 (en) 2010-06-16

Similar Documents

Publication Publication Date Title
Luo et al. High glucose inhibits myogenesis and induces insulin resistance by down-regulating AKT signaling
Jiao et al. Hepatic ERK activity plays a role in energy metabolism
Togliatto et al. Unacylated ghrelin induces oxidative stress resistance in a glucose intolerance and peripheral artery disease mouse model by restoring endothelial cell miR-126 expression
Seale et al. Prdm16 determines the thermogenic program of subcutaneous white adipose tissue in mice
Zhang et al. Long non‐coding RNA cardiac hypertrophy‐associated regulator governs cardiac hypertrophy via regulating miR‐20b and the downstream PTEN/AKT pathway
Lin et al. Canagliflozin alleviates LPS-induced acute lung injury by modulating alveolar macrophage polarization
Sabatini et al. Npas4 is a novel activity–regulated cytoprotective factor in pancreatic β-cells
Zhou et al. Omentin-1 protects against bleomycin-induced acute lung injury
Huang et al. Hypoxia-inducible factor 1α regulates the expression of the mitochondrial ATPase inhibitor protein (IF1) in rat liver
Zhang et al. Free fatty acids increase PGC-1α expression in isolated rat islets
US20090093434A1 (en) Use of eif-5a1 sirna to protect islets cells from apoptosis and to preserve their functionality
Wang et al. Pva-miR-252 participates in ammonia nitrogen-induced oxidative stress by modulating autophagy in Penaeus vannamei
Nagai et al. Rho-associated, coiled-coil–containing protein kinase 1 regulates development of diabetic kidney disease via modulation of fatty acid metabolism
He et al. Specific deletion of CDC42 in pancreatic β cells attenuates glucose-induced insulin expression and secretion in mice
Pan et al. Semaglutide alleviates inflammation-Induced endothelial progenitor cells injury by inhibiting MiR-155 expression in macrophage exosomes
He et al. Autophagy protects against high uric acid-induced hepatic insulin resistance
Zhu et al. Inhibition of Lincpint expression affects insulin secretion and apoptosis in mouse pancreatic β cells
Tyka et al. MCPIP1 is a novel link between diabetogenic conditions and impaired insulin secretory capacity
Garcia-Ocaña et al. Glucose mediated regulation of beta cell proliferation
Kong et al. Akt2 deficiency alleviates oxidative stress in the heart and liver via up-regulating SIRT6 during high-fat diet-induced obesity
Xie et al. Role of profilin-1 in vasculopathy induced by advanced glycation end products (AGEs)
Yorichika et al. The effects of Tel2 on cardiomyocyte survival
KR102413477B1 (en) Screening method for substance targeting USP21
CN113713107B (en) Application of miRNA552 cluster microRNA in treatment of glycolipid metabolic disease
JP5543774B2 (en) A novel method to prevent islet cells from being apoptotic during the donor harvest process

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880112165.0

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08798216

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2010521983

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 204072

Country of ref document: IL

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 1994/DELNP/2010

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 20107006218

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 584141

Country of ref document: NZ

Ref document number: 2008288988

Country of ref document: AU

Ref document number: 2008798216

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2700463

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2008288988

Country of ref document: AU

Date of ref document: 20080820

Kind code of ref document: A