WO2015196191A1 - Microrna biomarkers for traumatic brain injury and methods of use thereof - Google Patents

Microrna biomarkers for traumatic brain injury and methods of use thereof Download PDF

Info

Publication number
WO2015196191A1
WO2015196191A1 PCT/US2015/036925 US2015036925W WO2015196191A1 WO 2015196191 A1 WO2015196191 A1 WO 2015196191A1 US 2015036925 W US2015036925 W US 2015036925W WO 2015196191 A1 WO2015196191 A1 WO 2015196191A1
Authority
WO
WIPO (PCT)
Prior art keywords
mir
mirnas
subject
tbi
biological sample
Prior art date
Application number
PCT/US2015/036925
Other languages
French (fr)
Inventor
Radha K. Maheshwari
Nagaraja S. BALAKATHIRESAN
Manish BHOMIA
Kevin Ka-Wang Wang
Linda PAPA
Original Assignee
The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc.
University Of Florida Research Foundation, Inc.
Orlando Health, Inc., D/B/A Orlando Regional Medical Center
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 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc., University Of Florida Research Foundation, Inc., Orlando Health, Inc., D/B/A Orlando Regional Medical Center filed Critical The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc.
Publication of WO2015196191A1 publication Critical patent/WO2015196191A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/178Oligonucleotides characterized by their use miRNA, siRNA or ncRNA

Definitions

  • the present invention relates to methods of diagnosing traumatic brain injury (TBI) in a subject.
  • TBI traumatic brain injury
  • the present invention also relates to methods of monitoring the progression of the TBI in a subject.
  • Traumatic brain injury is a problem with epidemic magnitude involving both civilian, military service members and professional athletes.
  • TBI Traumatic brain injury
  • the economic burden of TBI in the United States is estimated to be $76.5 billion annually, in total lifetime direct medical costs and productivity losses.
  • Mild TBI also called concussion
  • mTBI Mild TBI
  • concussion accounts for more than 77 % of the total reported TBI cases in the United States. Among these cases it is estimated that around 40% of injuries are often ignored and do not seek medical attention.
  • mTBI is also a major cause of morbidity in the veterans returning from the recent wars with more than 20% of the veterans returning from the recent wars in Iraq and Afghanistan experienced a mTBI. Most of the symptoms associated with mTBI resolve within days or weeks of injury with substantial recovery in most cases. However, approximately, 10-20% of mTBI patients complain of prolonged problems and some experience symptoms lasting more than a year. mTBI can induce neurological, cognitive and behavioral changes in an individual.
  • the clinical symptoms may include headaches, sleep disturbance, impaired memory, anxiety and depression.
  • the accelerating and decelerating forces during the impact to the head also results in the injury to the white matter causing diffuse axonal injury.
  • Axonal injury may peak at 24 h post injury and can progress up to a year post injury. It is believed that this continuous progression may be a causative factor for the poor outcome post mTBI.
  • mTBI usually is a challenge for the clinicians to diagnose because of the lack of apparent signs of a brain injury.
  • mTBI is currently assessed using the Glasgow comma scale (GCS) which measure a score by assessing the eye, verbal, and motor response of the patient. GCS score and loss or alterations of consciousness are used to determine the severity of the injury.
  • GCS score can be of limited use in mTBI diagnosis due to the presence of polytrauma, alcohol abuse, use of sedatives and psychological stress.
  • Computed tomography and magnetic resonance imaging (MRI) are used to detect the extent of brain injury, however, in case of a concussion, CT and MRIs often fail to detect any specific injury lesion due to limited sensitivity and absence of micro-bleeds.
  • MRIs have become more sensitive than CT but due to their limited availability and the cost of the scan makes the utilization of this technique difficult for the acute stage diagnosis for both military and civilians.
  • Biomarkers in biofluids offer many advantages for mTBI diagnosis since they can be measured from the peripheral tissues such as blood, urine and saliva and can be easily quantitated using existing methods.
  • CSF cerebrospinal fluid
  • S- ⁇ S-100 calcium binding protein
  • GFAP glial fibrillary acidic protein
  • UCH-L1 Ubiquitin C-Terminal Hydrolase-Ll
  • MicroRNAs are small (19-28nt) endogenous RNA molecules that regulate protein synthesis at post transcriptional level. MiRNAs can be detected in serum and can be an indicator of disease pathology in the cell of origin including neuronal cells. This property of reflecting a diseased condition has recently gained attention towards miRNAs as biomarkers of central nervous system (CNS) pathology. Serum miRNAs are relatively stable and are resistant to repeated freeze thaw, enzymatic degradation and can survive variable pH conditions which make them a suitable biomarker candidate for mTBI.
  • CNS central nervous system
  • MiRNAs have been recently reported as specific and sensitive biomarkers of many CNS diseases.
  • the serum expression of miRNAs in response to a concussive mild injury in a closed head injury model was recently reported, and a signature of nine miRNAs was found to be modulated in serum immediately after the injury.
  • MiRNA modulation was also analyzed in a rodent model of traumatic stress, and a signature of 9 miRNAs was identified which were upregulated in serum and amygdala of the animals 2 weeks post exposure to traumatic stress.
  • miRNAs reported in this study did not have any similarities with the miRNAs reported for TBI studies, suggesting miRNA expression in serum may be a specific indicator of the altered physical state of the brain. There remains a need for a noninvasive, sensitive reliable test for diagnosis and monitoring TBI.
  • the present invention relates to methods of diagnosing traumatic brain injury (TBI) in a subject, the method comprising (a) determining a level(s) of one or more specific microRNAs (miRNAs) in a biological sample taken from the subject, and (b) comparing the determined level(s) of the one or more miRNAs against a level(s) of the same one or more miRNAs from a control subject determined not to be suffering from TBI, wherein an increase in the level(s) of the one or more miRNAs compared to level(s) of the one or more miRNAs from the control subject determined not to be suffering from TBI is indicative that the subject may be suffering from TBI.
  • TBI traumatic brain injury
  • the present invention also relates to methods of monitoring the progression of traumatic brain injury (TBI) in a subject, the method comprising (a) analyzing at least two biological samples from the subject taken at different time points to determine a level(s) of one or more specific miRNAs, and (b) comparing the level(s) of the one or more specific miRNAs over time to determine if the subject's level(s) of the one or more specific miRNAs is changing over time, wherein an increase in the level(s) of the one or more specific miRNAs over time is indicative that the subject's risk of suffering from TBI is increasing over time.
  • TBI traumatic brain injury
  • said miRNA is selected from the group consisting of miR-194, miR- 361, miR-625*, miR-1255B, miR-381, miR-425*, miR-638, miR-93, miR-328, miR-151-5p, miR-362-3p, miR-486, miR-942, miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR- 130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, and miR-491.
  • the TBI is mild TBI (mTBI) or severe TBI (sTBI).
  • the TBI is a closed head injury (CHI) or a blast-induced traumatic brain injury (bTBI).
  • the subject is human.
  • the biological sample is a serum and/or plasma sample.
  • the biological sample is taken from the subject more than one day, or more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14 days after the suspected traumatic episode.
  • the level(s) of one or more specific miRNAs are determined by a real time PCR.
  • the methods of diagnosing the TBI according to some embodiments of the present specification further comprise amplifying the miRNAs.
  • Figure 1 depicts miRNA specific validation assays in CSF samples of sTBI. Specific miRNA assays were performed for the five candidate miRNAs. Normalization was done with mir-202 which showed the least standard deviation and was selected as a normalizing control. Among the five tested, miRNAs, miR-328, miR-362-3p and miR-486 were significantly upregulated. Values are expressed as fold change+ SD over control in linear scale. Significance was calculated using paired student t test (p ⁇ 0.05).
  • the present invention relates to microRNA (miRNA) biomarkers from subjects with mild and severe traumatic brain injury (TBI), and their use thereof.
  • MiRNAs are small RNA molecules (e.g. 22 nucleotides long) and are often, but need not be, post-transcriptional regulators that bind to complementary sequences on target messenger RNA transcripts (mRNAs), usually resulting in translational repression and gene silencing.
  • mRNAs target messenger RNA transcripts
  • MiRNAs may serve as good biomarkers because they are highly stable in serum due to their ability to withstand repeated freeze thaw, enzymatic degradation, and extreme pH conditions.
  • miRNA includes human miRNAs, mature single stranded miRNAs, precursor miRNAs (pre-miR), and variants thereof, which may be naturally occurring.
  • miRNA also includes primary miRNA transcripts and duplex miRNAs.
  • the name of a specific miRNA refers to the mature miRNA.
  • miR-194 refers to a mature miRNA sequence derived from pre-miR-194.
  • miRNAs including human mature and precursor sequences
  • sequences for particular miRNAs are reported, for example, in miRBase:: Sequences Database on the web at: mirbase.org (version 20 released June 2013); Griffiths-Jones et al, Nucleic Acids Research, 2008, 36, Database Issue, D154-D158; Griffiths-Jones et al, Nucleic Acids Research, 2006, 34, Database Issue, D140-D144; Griffiths-Jones, Nucleic Acids Research, 2004, 32, Database Issue, D 109-D1 11.
  • a single precursor contains more than one mature miRNA sequence.
  • multiple precursor miRNAs contain the same mature sequence.
  • mature miRNAs have been re-named based on new scientific consensus. The skilled artisan will appreciate that scientific consensus regarding the precise nucleic acid sequence for a given miRNAs, in particular for mature forms of the miRNAs, may change with time.
  • the present invention relates to methods of diagnosing traumatic brain injury (TBI) in a subject.
  • the methods comprise (a) determining a level(s) of one or more miRNAs in a biological sample taken from the subject, and (b) comparing the determined level(s) of the one or more miRNAs against a level(s) of the same one or more miRNAs from a control subject determined not to be suffering from TBI.
  • An increase in the level(s) of the one or more miRNAs compared to level(s) of the one or more miRNAs from the control subject determined not to be suffering from TBI may be indicative that the subject may be suffering from TBI.
  • the present invention also relates to methods of monitoring the progression of traumatic brain injury (TBI) in a subject.
  • the method comprises (a) analyzing at least two biological samples from the subject taken at different time points to determine a level(s) of one or more specific miRNAs, and (b) comparing the level(s) of the one or more specific miRNAs over time to determine if the subject's level(s) of the one or more specific miRNAs is changing over time. An increase in the level(s) of the one or more specific miRNAs over time may be indicative that the subject's risk of suffering from TBI is increasing over time.
  • diagnosis includes making diagnostic or prognostic determinations or predictions of disease. In some instances, “diagnosing” includes identifying whether a subject has a disease such as TBI. Additionally, “diagnosing” includes distinguishing patients with mild TBI from patients having severe TBI. In other circumstances,
  • diagnosis includes determining the stage or aggressiveness of a disease state, or determining an appropriate treatment method for TBI.
  • the methods of the present inventions use miRNAs as markers for TBI.
  • miRNAs that are present at elevated levels in a biological sample (e.g. serum or plasma) from a subject with TBI are used as markers.
  • miRNAs that have reduced levels are used as markers.
  • more than one miRNA from the biological sample may be used as markers. When more than one miRNA biomarker is used, the miRNAs may all have elevated levels, all have reduced levels, or a mixture of miRNAs with elevated and reduced levels may be used.
  • an increase in the level(s) of the one or more miR As refers to an increase in the amount of a miRNA in a biological sample from a subject compared to the amount of the miRNA in the biological sample from a cohort or cohorts that do not have the TBI that the subject is being tested for. For instance, increased levels of miRNA in the biological sample indicate presence or prognosis for the TBI. In additional embodiments, certain miRNAs may be present in reduced levels in subjects with TBI. In some
  • the level of the miRNAs marker will be compared to a control to determine whether the level is decreased or increased.
  • the control may be, for example, miRNAs in a biological sample from a subject known to be free of TBI. In other embodiments, the control may be miRNAs from a non-serum sample like a tissue sample or a known amount of a synthetic RNA. In additional embodiments, the control may be miRNAs in a biological sample from the same subject at a different time.
  • said miRNA is selected from the group consisting of miR-194, miR- 361, miR-625*, miR-1255B, miR-381, miR-425*, miR-638, miR-93, miR-328, miR-151-5p, miR-362-3p, miR-486, miR-942, miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR- 130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, and miR-491. These miRNAs have elevated levels in serum from patients with TBI. These miRNAs may be used in accordance with the present inventions.
  • miRNAs may be useful for diagnosing TBI, including distinguishing mild and severe TBI.
  • these miRNA may be used to predict the aggressiveness or outcome of TBI.
  • said one or more miRNAs is selected from the group consisting of miR-194, miR-361, miR-625*, miR-1255B, miR- 381, miR-425*, miR-638, miR-93, miR-328, miR-151-5p, miR-362-3p, miR-486, miR-942. These miRNAs may be used to diagnose mild TBI.
  • said one or more miRNAs is selected from the group consisting of miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR-130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, miR-491, miR-328, miR-151-5p, miR-362-3p, miR-486, and miR-942. These miRNAs may be used to diagnose severe TBI. In another aspect, said one or more miRNAs is selected from the group consisting of miR-328, miR-151-5p, miR-362-3p, miR-486, and miR-942.
  • the miRNAs comprise at least miR-328, miR-362-3p and miR-486.
  • the methods may comprise assessing only miR-328, miR-362-3p and miR-486.
  • the methods comprise at least miR-328, miR-362-3p and miR-486, plus any one or more of miR-194, miR-361, miR-625*, miR-1255B, miR-381, miR-425*, miR-638, miR-93, miR-151-5p, miR-942, miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR-130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, and miR-491.
  • TBI may be classified as mild TBI or severe TBI.
  • the TBI is a closed head injury (CHI) or a blast-induced traumatic brain injury (bTBI).
  • CHI closed head injury
  • bTBI blast-induced traumatic brain injury
  • injury severity may be based on duration of loss of consciousness and/or coma rating scale or score, post-traumatic amnesia (PTA), and/or brain imaging results.
  • PTA post-traumatic amnesia
  • mild TBI may be characterized by brief loss of consciousness (e.g. a few seconds or minutes), PTA for less than 1 hour of the TBI, and normal brain imaging results.
  • a case of mild traumatic brain injury may be an occurrence of injury to the head resulting from blunt trauma or acceleration or deceleration forces with one or more of the following conditions attributable to the head injury during the surveillance period: (i) any period of observed or self-reported transient confusion, disorientation, or impaired consciousness; (ii) any period of observed or self-reported dysfunction of memory (amnesia) around the time of injury; (iii) Observed signs of other neurological or
  • neuropsychological dysfunction such as seizures acutely following head injury, irritability, lethargy, or vomiting following head injury among infants and very young children, and among older children and adults, headache, dizziness, irritability, fatigue, or poor
  • the subject is human or animal.
  • the biological samples described herein include, but is not limited to, blood, plasma, serum, urine, sputum, cerebrospinal fluid, milk, and ductal fluid samples.
  • the biological sample is a serum and/or plasma sample.
  • Serum is typically the fluid, non-cellular portion of coagulated blood.
  • Plasma is also a non-cellular blood sample, but unlike serum, plasma contains clotting factors.
  • serum or plasma samples may be obtained from a human subject previously screened for TBI using other diagnostic methods.
  • Additional embodiments include measuring miRNA in samples from subjects previously or currently undergoing treatment for TBI.
  • the volume of plasma or serum obtained and used in the methods described herein may be varied depending upon clinical intent.
  • One of skill in the art may recognize that many methods exist for obtaining and preparing serum samples. Generally, blood is drawn into a collection tube using standard methods and allowed to clot. The serum is then separated from the cellular portion of the coagulated blood. In methods according to some embodiments of the present inventions, clotting activators such as silica particles are added to the blood collection tube. In other methods, the blood is not treated to facilitate clotting. Blood collection tubes are
  • Dickenson Vacutainer® tubes SSTTM, glass serum tubes, or plastic serum tubes.
  • the blood is collected by venipuncture and processed within three hours after drawing to minimize hemolysis and minimize the release of miRNAs from intact cells in the blood.
  • blood is kept on ice until use.
  • the blood may be fractionated by centrifugation to remove cellular components.
  • centrifugation to prepare serum can be at a speed of at least 500, 1000, 2000, 3000, 4000, or 5000xG.
  • the blood can be incubated for at least 10, 20, 30, 40, 50, 60, 90, 120, or 150 minutes to allow clotting. In other embodiments, the blood is incubated for at most 3 hours.
  • the blood is not permitted to coagulate prior to separation of the cellular and acellular components. Serum or plasma may be frozen after separation from the cellular portion of blood until further assayed.
  • RNA may be extracted from serum or plasma and purified using methods known in the art. Many methods are known for isolating total RNA, or to specifically extract small RNAs, including miRNAs.
  • the RNA may be extracted using commercially-available kits (e.g., Perfect RNA Total RNA Isolation Kit, Five Prime-Three Prime, Inc.; mirVanaTM kits, Ambion, Inc.).
  • RNA extraction methods previously published for the extraction of mammalian intracellular RNA or viral RNA may be adapted, either as published or with modification, for extraction of RNA from plasma and serum.
  • RNA may be extracted from plasma or serum using silica particles, glass beads, or diatoms, as in the method or adaptations described in U.S.
  • the biological sample may be collected from a subject more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, or 14 days after a suspected traumatic episode. In another aspect, the biological sample may be collected from a subject less than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, or 15 days after a suspected traumatic episode.
  • the level(s) of one or more specific miR As are determined by a real time PCR. In some embodiments, the methods of the present inventions comprise amplifying the miRNAs.
  • miRNAs are amplified prior to measurement.
  • the level of miRNAs is measured during the amplification process.
  • the miRN As is not amplified prior to measurement.
  • nucleic acid polymerization and amplification techniques include reverse transcription (RT), polymerase chain reaction (PCR), real-time PCR (quantitative PCR (q-PCR)), nucleic acid sequence-base amplification (NA8BA), ligase chain reaction, multiplex ligatable probe amplification, invader technology (Third Wave), rolling circle amplification, in vitro transcription (IVT), strand displacement amplification, transcription-mediated amplification (TMA), RNA (Eberwine) amplification, and other methods that are known to persons skilled in the art.
  • more than one amplification method is used, such as reverse transcription followed by real time quantitative PCR (qRT-PCR) (Chen et al,, Nucleic Acids Research, 33(20):el79 (2005)).
  • a typical PCR reaction includes multiple amplification steps, or cycles that selectively amplify target nucleic acid species: a denaturing step in which a target nucleic acid is denatured; an annealing step in which a set of PCR primers (forward and reverse primers) anneal to complementary UNA strands; and an elongation step in which a thermostable DNA polymerase elongates the primers. By repeating these steps multiple times, a DNA fragment is amplified to produce an amplicon, corresponding to the target DNA sequence.
  • Typical PCR reactions include 20 or more cycles of denaturation, annealing, and elongation.
  • annealing and elongation steps can be performed concurrently, in which case the cycle contains only two steps.
  • a reverse transcription reaction (which produces a complementary cDNA sequence) may be performed prior to PCR reactions.
  • Reverse transcription reactions include the use of, e.g., a RNA-based DNA polymerase (reverse transcriptase) and a primer.
  • a set of primers is used for each target sequence.
  • the lengths of the primers depends on many factors, including, but not limited to, the desired hybridization temperature between the primers, the target nucleic acid sequence, and the complexity of the different target nucleic acid sequences to be amplified.
  • a primer is about 15 to about 35 nucleotides in length. In other embodiments, a primer is equal to or fewer than 15, 20, 25, 30, or 35 nucleotides in length. In additional embodiments, a primer is at least 35 nucleotides in length.
  • a forward primer can comprise at least one sequence that anneals to a miRNA biomarker and alternatively can comprise an additional 5' non-complementary region.
  • a reverse primer can be designed to anneal to the complement of a reverse transcribed miRNAs.
  • the reverse primer may be independent of the miRNA biomarker sequence, and multiple miRNA biomarkers may be amplified using the same reverse primer.
  • a reverse primer may be specific for a miRNA biomarker.
  • two or more miRNAs are amplified in a single reaction volume.
  • One aspect includes multiplex q-PCR, such as Real Time quantitative PGR (qRT- PCR), which enables simultaneous amplification and quantification of at least two miRNAs of interest in one reaction volume by using more than one pair of primers and/or more than one probe.
  • the primer pairs comprise at least one amplification primer that uniquely binds each miRNA, and the probes are labeled such that they are distinguishable from one another, thus allowing simultaneous quantification of multiple miRNAs.
  • Multiplex qRT-PCR has research and diagnostic uses, including but not limited to detection of miRNAs for diagnostic, prognostic, and therapeutic applications.
  • the qRT-PCR reaction may further be combined with the reverse transcription reaction by including both a reverse transcriptase and a DN A-based thermostable DNA polymerase.
  • a "hot start” approach may be used to maximize assay performance (U.S. Pat. Nos. 5,41 1,876 and 5,985,619).
  • the components for a reverse transcriptase reaction and a PGR reaction may be sequestered using one or more thermoactivation methods or chemical alteration to improve polymerization efficiency (U.S. Pat. Nos. 5,550,044, 5,413,924, and 6,403,341 ).
  • labels, dyes, or labeled probes and/or primers are used to detect amplified or unamplified miRNAs.
  • detection methods are appropriate based on the sensitivity of the detection method and the abundance of the target.
  • amplification may or may not be required prior to detection.
  • miRNA amplification is preferred.
  • a probe or primer may include Watson-Crick bases or modified bases.
  • Modified bases include, but are not limited to, the AEGIS bases (from Eragen Biosciences), which have been described, e.g., in U.S. Pat. Nos. 5,432,272, 5,965,364, and 6,001,983.
  • bases are joined by a natural phosphodiester bond or a different chemical linkage.
  • Different chemical linkages include, but are not limited to, a peptide bond or a Locked Nucleic Acid (LISA) linkage, which is described, e.g., in U.S. Pat, No. 7,060,809.
  • oligonucleotide probes or primers present in an amplification reaction are suitable for monitoring the amount of amplification product produced as a function of time.
  • probes having different single stranded versus double stranded character are used to detect the nucleic acid.
  • Probes include, but are not limited to, the 5'-exonuclease assay (e.g., TaqManTM) probes (see U.S. Pat. No. 5,538,848), stem-loop molecular beacons (see, e.g., U.S. Pat. Nos.
  • stemfess or lineal- beacons see, e.g., WO 9921881, U.S. Pat. Nos. 6,485,901 and 6,649,349), peptide nucleic acid (PNA) Molecular Beacons (see, e.g., U.S. Pat. Nos. 6,355,421 and 6,593,091 ), linear PNA beacons (see, e.g. U.S. Pat. No. 6,329,144), non-FRET probes (see, e.g., U.S. Pat. No. 6, 150,097), SunriseTM/AmplifiuorBTMprobes (see, e.g., U.S.
  • one or more of the primers in an amplification reaction can include a label
  • different probes or primers comprise detectable labels that are distinguishable from one another, in some embodiments a nucleic acid, such as the probe or primer, may be labeled with two or more distinguishable labels.
  • a label is attached to one or more probes and has one or more of the following properties: (i) provides a detectable signal; (ii) interacts with a second label to modify the detectable signal provided by the second label, e.g., FRET (Fluorescent
  • a binding complex or affinity set e.g., affinity, antibody-antigen, ionic complexes, hapten-ligand (e.g., biotin-avidin).
  • use of labels can be accomplished using any one of a large number of known techniques employing known labels, linkages, linking groups, reagents, reaction conditions, and analysis and purification methods.
  • MiRNAs can be deiected by direct or indirect methods, in a direct detection method, one or more miRNAs are detected by a detectable label that is linked to a nucleic acid molecule. In such methods, the miRNAs may be labeled prior to binding to the probe.
  • binding is detected by screening for the labeled miRNAs that is bound to the probe.
  • the probe is optionally linked to a bead in the reaction volume.
  • nucleic acids are detected by direct binding with a labeled probe, and the probe is subsequently detected.
  • the nucleic acids such as amplified miRNAs, are detected using FlexMAP Microspheres (Luminex) conjugated with probes to capture the desired nucleic acids.
  • Some methods may involve detection with polynucleotide probes modified with fluorescent labels or branched DNA (bDNA) detection, for example.
  • bDNA branched DNA
  • nucleic acids are detected by indirect detection methods.
  • a biotinylated probe may be combined with a streptavidin-conjugated dye to detect the bound nucleic acid.
  • the streptavidin molecule binds a biotin label on amplified miRNAs, and the bound miRNA is detected by detecting the dye molecule attached to the streptavidin molecule.
  • the streptavidin-conjugated dye molecule comprises
  • Labels include, but are not limited to: light-emitting, light-scattering, and light- absorbing compounds which generate or quench a detectable fluorescent, chemiiuminescent, or bioluminescent signal (see, e.g., Kricka, L., Nonisotopic DNA Probe Techniquies, Academic Press, San Diego (1992) and Ganman A ., on-Radioactive Labeling, Academic Press (1997).
  • Fluorescent reporter dyes useful as labels include, but are not limited to, fluoresceins (see, e.g., U.S. Pat. Nos.
  • benzophenoxazincs see, e.g., U.S. Pat. No. 6, 140,500
  • energy-transfer fluorescent dyes comprising pairs of donors and acceptors (see, e.g., U.S. Pat. Nos, 5,863,727; 5,800,996; and 5,945,526), and cyanines (see, e.g., WO 9745539), iissamine, phycoerythrin, Cy2, Cy3, Cy3.5, Cy5, Cy5.5, Cy7, FluorX (Amersham), Alexa 350, Alexa 430, AMCA, BOD1PY 630/650, BODTPY 650/665, BODIPY-FL, BODIPY-R6G, BODIPY-TMR, BODIPY-TRX, Cascade Blue, Cy3, Cy5, 6-FAM, Fluorescein Isothiocyaoate, HEX, 6-JOE, Oregon Green 488, Oregon Green 500, Oregon Green 514,
  • fluorescein dyes include, but are not limited to, 6-carboxyfluorescein, 2',4',1 ,4,- teirachlorofluorescein and 2',4',5',7', 1 ,4-hexachlorofluorescein.
  • the fluorescent label is selected from SYBR-Green, 6-carboxyfluorescein ("FAM”), TET, ROX, VICTM, and JOE.
  • FAM 6-carboxyfluorescein
  • TET 6-carboxyfluorescein
  • ROX ROX
  • VICTM VICTM
  • JOE JOE
  • labels are different fluorophores capable of emitting light at different, spectrally-resolvable wavelengths (e.g., 4-differently colored fluorophores); certain such labeled probes are known in the art and described above, and in U.S. Pat. No.
  • a dual labeled fluorescent probe that includes a reporter fluorophore and a quencher fluorophore is used in some embodiments. It will be appreciated that pairs of fluorophores are chosen that have distinct emission spectra so that they can be easily dis inguished.
  • labels are hybridization-stabilizing moieties which serve to enhance, stabilize, or influence hybridization of duplexes, e.g., intercalators and intercalating dyes (including, but not limited to, ethidium bromide and SYBR-Green), minor-groove binders, and cross-linking functional groups (see, e.g., Blackburn et ak, eds. "DNA and RNA Structure” in Nucleic Acids in Chemistry and Biology (1996)).
  • intercalators and intercalating dyes including, but not limited to, ethidium bromide and SYBR-Green
  • minor-groove binders include, but not limited to, ethidium bromide and SYBR-Green
  • cross-linking functional groups see, e.g., Blackburn et ak, eds. "DNA and RNA Structure” in Nucleic Acids in Chemistry and Biology (1996)).
  • methods relying on hybridization and/or ligation to quantify miRNAs may be used, including oligonucleotide ligation (OLA) methods and methods that allow a distinguishable probe that hybridizes to the target nucleic acid sequence to be separated from an unbound probe.
  • OLA oligonucleotide ligation
  • HARP-like probes as disclosed in U.S. Publication No, 2006/0078894 may be used to measure the amount of miRNAs.
  • the probe after hybridization between a probe and the targeted nucleic acid, the probe is modified to distinguish the hybridized probe from the unhybridized probe. Thereafter, the probe may be amplified and/or detected.
  • a probe inactivation region comprises a subset of nucleotides within the target hybridization region of the probe.
  • a post-hybridization probe inactivation step is carried out using an agent which is able to distinguish between a HARP probe that is hybridized to its targeted nucleic acid sequence and the corresponding unhybridized HARP probe.
  • the agent is able to inactivate or modify the unhybridized HARP probe such that it cannot be amplified.
  • a probe ligation reaction may be used to quantify miRNAs.
  • MLPA Multiplex Ligation-dependent Probe Amplification
  • pairs of probes which hybridize immediately adjacent to each other on the target nucleic acid are li gated to each other only in the presence of the target nucleic acid.
  • MLPA probes have flanking PCR primer binding sites. MLPA probes can only be amplified if they have been li gated, thus allowing for detection and quantification of miRNA biomarkers.
  • RNA profiling was performed using Taqman low density array platform for human miRNAs followed by data analysis. RealTime StatMiner® from Integromics® bioinformatics tool was used for the identification of significantly altered miRNA levels in the serum samples.
  • Table 1 Altered miRNAs as biomarkers of mild TBI.
  • Table 2 Altered miRNAs as biomarkers of severe TBI.
  • miRNA biomarkers as shown in Table 3 were found to be present in the samples from the subjects with mTBI and sTBI, but not in the samples from the subjects with the orthopedic injury. Comparison of these 5 miRNAs with serum miRNA profiles of animal TBI models revealed similar miRNAs between human and animal serum post injury. Table 3 shows the potential biomarker candidates in mild and severe TBI along with their normalized fold changes indicating their level of expression.
  • Table 3 MiRNA biomarkers for mild and severe TBI.

Abstract

The present invention relates to methods of diagnosing traumatic brain injury (TBI) in a subject. The present invention also relates to methods of monitoring the progression of the TBI in a subject.

Description

MICRORNA BIOMARKERS FOR TRAUMATIC BRAIN INJURY
AND METHODS OF USE THEREOF
Field of Invention
[0001] The present invention relates to methods of diagnosing traumatic brain injury (TBI) in a subject. The present invention also relates to methods of monitoring the progression of the TBI in a subject.
Background of the Invention
[0002] Traumatic brain injury (TBI) is a problem with epidemic magnitude involving both civilian, military service members and professional athletes. In the United States, more than 1.3 million emergency room visits account for TBI and is a cause of almost a third of all injury related deaths. The economic burden of TBI in the United States is estimated to be $76.5 billion annually, in total lifetime direct medical costs and productivity losses.
[0003] Mild TBI (mTBI), also called concussion, accounts for more than 77 % of the total reported TBI cases in the United States. Among these cases it is estimated that around 40% of injuries are often ignored and do not seek medical attention. mTBI is also a major cause of morbidity in the veterans returning from the recent wars with more than 20% of the veterans returning from the recent wars in Iraq and Afghanistan experienced a mTBI. Most of the symptoms associated with mTBI resolve within days or weeks of injury with substantial recovery in most cases. However, approximately, 10-20% of mTBI patients complain of prolonged problems and some experience symptoms lasting more than a year. mTBI can induce neurological, cognitive and behavioral changes in an individual. The clinical symptoms may include headaches, sleep disturbance, impaired memory, anxiety and depression. The accelerating and decelerating forces during the impact to the head also results in the injury to the white matter causing diffuse axonal injury. Axonal injury may peak at 24 h post injury and can progress up to a year post injury. It is believed that this continuous progression may be a causative factor for the poor outcome post mTBI.
[0004] mTBI usually is a challenge for the clinicians to diagnose because of the lack of apparent signs of a brain injury. mTBI is currently assessed using the Glasgow comma scale (GCS) which measure a score by assessing the eye, verbal, and motor response of the patient. GCS score and loss or alterations of consciousness are used to determine the severity of the injury. The GCS score can be of limited use in mTBI diagnosis due to the presence of polytrauma, alcohol abuse, use of sedatives and psychological stress. Computed tomography (CT) and magnetic resonance imaging (MRI) are used to detect the extent of brain injury, however, in case of a concussion, CT and MRIs often fail to detect any specific injury lesion due to limited sensitivity and absence of micro-bleeds. With new technological
advancements, MRIs have become more sensitive than CT but due to their limited availability and the cost of the scan makes the utilization of this technique difficult for the acute stage diagnosis for both military and civilians.
[0005] Biomarkers in biofluids offer many advantages for mTBI diagnosis since they can be measured from the peripheral tissues such as blood, urine and saliva and can be easily quantitated using existing methods. Several protein markers in serum and cerebrospinal fluid (CSF) like S-100 calcium binding protein (S-ΙΟΟβ), glial fibrillary acidic protein (GFAP) and Ubiquitin C-Terminal Hydrolase-Ll (UCH-L1) have been extensively studied for their utility as biomarkers for mild to severe TBI (sTBI). However, most of the protein biomarkers studied have relatively less sensitivity for mTBI with no intracranial lesions. Combinations of more than one protein biomarkers for mTBI diagnosis have been recently studied, and these show better diagnostic accuracy in comparison to single markers. Despite extensive studies most of the protein markers are in preclinical testing and none of the markers are available for clinical use.
[0006] MicroRNAs (miRNA) are small (19-28nt) endogenous RNA molecules that regulate protein synthesis at post transcriptional level. MiRNAs can be detected in serum and can be an indicator of disease pathology in the cell of origin including neuronal cells. This property of reflecting a diseased condition has recently gained attention towards miRNAs as biomarkers of central nervous system (CNS) pathology. Serum miRNAs are relatively stable and are resistant to repeated freeze thaw, enzymatic degradation and can survive variable pH conditions which make them a suitable biomarker candidate for mTBI.
[0007] MiRNAs have been recently reported as specific and sensitive biomarkers of many CNS diseases. The serum expression of miRNAs in response to a concussive mild injury in a closed head injury model was recently reported, and a signature of nine miRNAs was found to be modulated in serum immediately after the injury. MiRNA modulation was also analyzed in a rodent model of traumatic stress, and a signature of 9 miRNAs was identified which were upregulated in serum and amygdala of the animals 2 weeks post exposure to traumatic stress. Interestingly, miRNAs reported in this study did not have any similarities with the miRNAs reported for TBI studies, suggesting miRNA expression in serum may be a specific indicator of the altered physical state of the brain. There remains a need for a noninvasive, sensitive reliable test for diagnosis and monitoring TBI.
Summary of the Invention
[0008] In one aspect, the present invention relates to methods of diagnosing traumatic brain injury (TBI) in a subject, the method comprising (a) determining a level(s) of one or more specific microRNAs (miRNAs) in a biological sample taken from the subject, and (b) comparing the determined level(s) of the one or more miRNAs against a level(s) of the same one or more miRNAs from a control subject determined not to be suffering from TBI, wherein an increase in the level(s) of the one or more miRNAs compared to level(s) of the one or more miRNAs from the control subject determined not to be suffering from TBI is indicative that the subject may be suffering from TBI.
[0009] In another aspect, the present invention also relates to methods of monitoring the progression of traumatic brain injury (TBI) in a subject, the method comprising (a) analyzing at least two biological samples from the subject taken at different time points to determine a level(s) of one or more specific miRNAs, and (b) comparing the level(s) of the one or more specific miRNAs over time to determine if the subject's level(s) of the one or more specific miRNAs is changing over time, wherein an increase in the level(s) of the one or more specific miRNAs over time is indicative that the subject's risk of suffering from TBI is increasing over time.
[0010] In one aspect, said miRNA is selected from the group consisting of miR-194, miR- 361, miR-625*, miR-1255B, miR-381, miR-425*, miR-638, miR-93, miR-328, miR-151-5p, miR-362-3p, miR-486, miR-942, miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR- 130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, and miR-491. In another aspect, the TBI is mild TBI (mTBI) or severe TBI (sTBI). In another aspect, the TBI is a closed head injury (CHI) or a blast-induced traumatic brain injury (bTBI). In another aspect, the subject is human. In another aspect, the biological sample is a serum and/or plasma sample. In another aspect, the biological sample is taken from the subject more than one day, or more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14 days after the suspected traumatic episode. [0011] In another aspect, the level(s) of one or more specific miRNAs are determined by a real time PCR. The methods of diagnosing the TBI according to some embodiments of the present specification further comprise amplifying the miRNAs.
Brief Description of the Drawings
[0012] Figure 1 depicts miRNA specific validation assays in CSF samples of sTBI. Specific miRNA assays were performed for the five candidate miRNAs. Normalization was done with mir-202 which showed the least standard deviation and was selected as a normalizing control. Among the five tested, miRNAs, miR-328, miR-362-3p and miR-486 were significantly upregulated. Values are expressed as fold change+ SD over control in linear scale. Significance was calculated using paired student t test (p<0.05).
Detailed Description of the Invention
[0013] The present invention relates to microRNA (miRNA) biomarkers from subjects with mild and severe traumatic brain injury (TBI), and their use thereof. MiRNAs are small RNA molecules (e.g. 22 nucleotides long) and are often, but need not be, post-transcriptional regulators that bind to complementary sequences on target messenger RNA transcripts (mRNAs), usually resulting in translational repression and gene silencing. MiRNAs may serve as good biomarkers because they are highly stable in serum due to their ability to withstand repeated freeze thaw, enzymatic degradation, and extreme pH conditions. As used herein, the term "microRNA" (miRNA) includes human miRNAs, mature single stranded miRNAs, precursor miRNAs (pre-miR), and variants thereof, which may be naturally occurring. In some instances, the term "miRNA" also includes primary miRNA transcripts and duplex miRNAs. Unless otherwise noted, when used herein, the name of a specific miRNA refers to the mature miRNA. For example, miR-194 refers to a mature miRNA sequence derived from pre-miR-194. The sequences for particular miRNAs, including human mature and precursor sequences, are reported, for example, in miRBase:: Sequences Database on the web at: mirbase.org (version 20 released June 2013); Griffiths-Jones et al, Nucleic Acids Research, 2008, 36, Database Issue, D154-D158; Griffiths-Jones et al, Nucleic Acids Research, 2006, 34, Database Issue, D140-D144; Griffiths-Jones, Nucleic Acids Research, 2004, 32, Database Issue, D 109-D1 11. For certain miRNAs, a single precursor contains more than one mature miRNA sequence. In other instances, multiple precursor miRNAs contain the same mature sequence. In some instances, mature miRNAs have been re-named based on new scientific consensus. The skilled artisan will appreciate that scientific consensus regarding the precise nucleic acid sequence for a given miRNAs, in particular for mature forms of the miRNAs, may change with time.
[0014] In another aspect, the present invention relates to methods of diagnosing traumatic brain injury (TBI) in a subject. In some embodiments, the methods comprise (a) determining a level(s) of one or more miRNAs in a biological sample taken from the subject, and (b) comparing the determined level(s) of the one or more miRNAs against a level(s) of the same one or more miRNAs from a control subject determined not to be suffering from TBI. An increase in the level(s) of the one or more miRNAs compared to level(s) of the one or more miRNAs from the control subject determined not to be suffering from TBI may be indicative that the subject may be suffering from TBI.
[0015] In another aspect, the present invention also relates to methods of monitoring the progression of traumatic brain injury (TBI) in a subject. In some embodiments, the method comprises (a) analyzing at least two biological samples from the subject taken at different time points to determine a level(s) of one or more specific miRNAs, and (b) comparing the level(s) of the one or more specific miRNAs over time to determine if the subject's level(s) of the one or more specific miRNAs is changing over time. An increase in the level(s) of the one or more specific miRNAs over time may be indicative that the subject's risk of suffering from TBI is increasing over time.
[0016] The term "diagnosing" includes making diagnostic or prognostic determinations or predictions of disease. In some instances, "diagnosing" includes identifying whether a subject has a disease such as TBI. Additionally, "diagnosing" includes distinguishing patients with mild TBI from patients having severe TBI. In other circumstances,
"diagnosing" includes determining the stage or aggressiveness of a disease state, or determining an appropriate treatment method for TBI.
[0017] In some embodiments, the methods of the present inventions use miRNAs as markers for TBI. In some embodiments, miRNAs that are present at elevated levels in a biological sample (e.g. serum or plasma) from a subject with TBI are used as markers. In other embodiments, miRNAs that have reduced levels are used as markers. In some embodiments, more than one miRNA from the biological sample may be used as markers. When more than one miRNA biomarker is used, the miRNAs may all have elevated levels, all have reduced levels, or a mixture of miRNAs with elevated and reduced levels may be used. [0018] The term "an increase in the level(s) of the one or more miR As" refers to an increase in the amount of a miRNA in a biological sample from a subject compared to the amount of the miRNA in the biological sample from a cohort or cohorts that do not have the TBI that the subject is being tested for. For instance, increased levels of miRNA in the biological sample indicate presence or prognosis for the TBI. In additional embodiments, certain miRNAs may be present in reduced levels in subjects with TBI. In some
embodiments, the level of the miRNAs marker will be compared to a control to determine whether the level is decreased or increased. The control may be, for example, miRNAs in a biological sample from a subject known to be free of TBI. In other embodiments, the control may be miRNAs from a non-serum sample like a tissue sample or a known amount of a synthetic RNA. In additional embodiments, the control may be miRNAs in a biological sample from the same subject at a different time.
[0019] In one aspect, said miRNA is selected from the group consisting of miR-194, miR- 361, miR-625*, miR-1255B, miR-381, miR-425*, miR-638, miR-93, miR-328, miR-151-5p, miR-362-3p, miR-486, miR-942, miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR- 130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, and miR-491. These miRNAs have elevated levels in serum from patients with TBI. These miRNAs may be used in accordance with the present inventions. These miRNAs may be useful for diagnosing TBI, including distinguishing mild and severe TBI. In addition, these miRNA may be used to predict the aggressiveness or outcome of TBI. In another aspect, said one or more miRNAs is selected from the group consisting of miR-194, miR-361, miR-625*, miR-1255B, miR- 381, miR-425*, miR-638, miR-93, miR-328, miR-151-5p, miR-362-3p, miR-486, miR-942. These miRNAs may be used to diagnose mild TBI. In another aspect, said one or more miRNAs is selected from the group consisting of miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR-130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, miR-491, miR-328, miR-151-5p, miR-362-3p, miR-486, and miR-942. These miRNAs may be used to diagnose severe TBI. In another aspect, said one or more miRNAs is selected from the group consisting of miR-328, miR-151-5p, miR-362-3p, miR-486, and miR-942.
[0020] In another aspect, the miRNAs comprise at least miR-328, miR-362-3p and miR-486. For example, the methods may comprise assessing only miR-328, miR-362-3p and miR-486. In another embodiment, the methods comprise at least miR-328, miR-362-3p and miR-486, plus any one or more of miR-194, miR-361, miR-625*, miR-1255B, miR-381, miR-425*, miR-638, miR-93, miR-151-5p, miR-942, miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR-130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, and miR-491.
[0021] In another aspect, TBI may be classified as mild TBI or severe TBI. In some embodiments, the TBI is a closed head injury (CHI) or a blast-induced traumatic brain injury (bTBI).
[0022] In one aspect, injury severity may be based on duration of loss of consciousness and/or coma rating scale or score, post-traumatic amnesia (PTA), and/or brain imaging results. In some cases, mild TBI may be characterized by brief loss of consciousness (e.g. a few seconds or minutes), PTA for less than 1 hour of the TBI, and normal brain imaging results. In additional embodiments, a case of mild traumatic brain injury may be an occurrence of injury to the head resulting from blunt trauma or acceleration or deceleration forces with one or more of the following conditions attributable to the head injury during the surveillance period: (i) any period of observed or self-reported transient confusion, disorientation, or impaired consciousness; (ii) any period of observed or self-reported dysfunction of memory (amnesia) around the time of injury; (iii) Observed signs of other neurological or
neuropsychological dysfunction, such as seizures acutely following head injury, irritability, lethargy, or vomiting following head injury among infants and very young children, and among older children and adults, headache, dizziness, irritability, fatigue, or poor
concentration, when identified soon after injury; and/or (iv) any period of observed or self- reported loss of consciousness lasting 30 minutes or less. In other cases, severe TBI may be characterized by loss of consciousness or coma for more than 24 hours, PTA for more than 24 hours of the TBI, and/or abnormal brain imaging results.
[0023] In another aspect, the subject is human or animal. In another aspect, the biological samples described herein include, but is not limited to, blood, plasma, serum, urine, sputum, cerebrospinal fluid, milk, and ductal fluid samples. In some embodiments, the biological sample is a serum and/or plasma sample. Serum is typically the fluid, non-cellular portion of coagulated blood. Plasma is also a non-cellular blood sample, but unlike serum, plasma contains clotting factors. In some embodiments, serum or plasma samples may be obtained from a human subject previously screened for TBI using other diagnostic methods.
Additional embodiments include measuring miRNA in samples from subjects previously or currently undergoing treatment for TBI. The volume of plasma or serum obtained and used in the methods described herein may be varied depending upon clinical intent. [0024] One of skill in the art may recognize that many methods exist for obtaining and preparing serum samples. Generally, blood is drawn into a collection tube using standard methods and allowed to clot. The serum is then separated from the cellular portion of the coagulated blood. In methods according to some embodiments of the present inventions, clotting activators such as silica particles are added to the blood collection tube. In other methods, the blood is not treated to facilitate clotting. Blood collection tubes are
commercially available from many sources and in a variety of formats (e.g., Becton
Dickenson Vacutainer® tubes— SST™, glass serum tubes, or plastic serum tubes).
[0025] In some embodiments, the blood is collected by venipuncture and processed within three hours after drawing to minimize hemolysis and minimize the release of miRNAs from intact cells in the blood. In some methods, blood is kept on ice until use. The blood may be fractionated by centrifugation to remove cellular components. In some embodiments, centrifugation to prepare serum can be at a speed of at least 500, 1000, 2000, 3000, 4000, or 5000xG. In certain embodiments, the blood can be incubated for at least 10, 20, 30, 40, 50, 60, 90, 120, or 150 minutes to allow clotting. In other embodiments, the blood is incubated for at most 3 hours. When using plasma, the blood is not permitted to coagulate prior to separation of the cellular and acellular components. Serum or plasma may be frozen after separation from the cellular portion of blood until further assayed.
[0026] Before performing the methods according to the present inventions, RNA may be extracted from serum or plasma and purified using methods known in the art. Many methods are known for isolating total RNA, or to specifically extract small RNAs, including miRNAs. The RNA may be extracted using commercially-available kits (e.g., Perfect RNA Total RNA Isolation Kit, Five Prime-Three Prime, Inc.; mirVana™ kits, Ambion, Inc.). Alternatively, RNA extraction methods previously published for the extraction of mammalian intracellular RNA or viral RNA may be adapted, either as published or with modification, for extraction of RNA from plasma and serum. RNA may be extracted from plasma or serum using silica particles, glass beads, or diatoms, as in the method or adaptations described in U.S.
Publication No. 2008/0057502.
[0027] In another aspect, the biological sample may be collected from a subject more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, or 14 days after a suspected traumatic episode. In another aspect, the biological sample may be collected from a subject less than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, or 15 days after a suspected traumatic episode. [0028] In another aspect, the level(s) of one or more specific miR As are determined by a real time PCR. In some embodiments, the methods of the present inventions comprise amplifying the miRNAs.
[0029] Many methods of measuring the levels or amounts of miRNAs are contemplated. Any reliable, sensitive, and specific method may be used. In some embodiments, the miRNAs are amplified prior to measurement. In other embodiments, the level of miRNAs is measured during the amplification process. In still other methods, the miRN As is not amplified prior to measurement.
[0030] Many methods exist for amplifying miRNA nucleic acid sequences such as mature miRNAs, primary miRNAs and precursor miRNAs. Suitable nucleic acid polymerization and amplification techniques include reverse transcription (RT), polymerase chain reaction (PCR), real-time PCR (quantitative PCR (q-PCR)), nucleic acid sequence-base amplification (NA8BA), ligase chain reaction, multiplex ligatable probe amplification, invader technology (Third Wave), rolling circle amplification, in vitro transcription (IVT), strand displacement amplification, transcription-mediated amplification (TMA), RNA (Eberwine) amplification, and other methods that are known to persons skilled in the art. In certain embodiments, more than one amplification method is used, such as reverse transcription followed by real time quantitative PCR (qRT-PCR) (Chen et al,, Nucleic Acids Research, 33(20):el79 (2005)).
[0031] A typical PCR reaction includes multiple amplification steps, or cycles that selectively amplify target nucleic acid species: a denaturing step in which a target nucleic acid is denatured; an annealing step in which a set of PCR primers (forward and reverse primers) anneal to complementary UNA strands; and an elongation step in which a thermostable DNA polymerase elongates the primers. By repeating these steps multiple times, a DNA fragment is amplified to produce an amplicon, corresponding to the target DNA sequence. Typical PCR reactions include 20 or more cycles of denaturation, annealing, and elongation. In many cases, the annealing and elongation steps can be performed concurrently, in which case the cycle contains only two steps. Since mature miRNAs are single-stranded, a reverse transcription reaction (which produces a complementary cDNA sequence) may be performed prior to PCR reactions. Reverse transcription reactions include the use of, e.g., a RNA-based DNA polymerase (reverse transcriptase) and a primer.
[0032] in PCR and q-PCR methods, for example, a set of primers is used for each target sequence. In certain embodiments, the lengths of the primers depends on many factors, including, but not limited to, the desired hybridization temperature between the primers, the target nucleic acid sequence, and the complexity of the different target nucleic acid sequences to be amplified. In certain embodiments, a primer is about 15 to about 35 nucleotides in length. In other embodiments, a primer is equal to or fewer than 15, 20, 25, 30, or 35 nucleotides in length. In additional embodiments, a primer is at least 35 nucleotides in length.
[0033] In a further aspect, a forward primer can comprise at least one sequence that anneals to a miRNA biomarker and alternatively can comprise an additional 5' non-complementary region. In another aspect, a reverse primer can be designed to anneal to the complement of a reverse transcribed miRNAs. The reverse primer may be independent of the miRNA biomarker sequence, and multiple miRNA biomarkers may be amplified using the same reverse primer. Alternatively, a reverse primer may be specific for a miRNA biomarker.
[0034] In some embodiments, two or more miRNAs are amplified in a single reaction volume. One aspect includes multiplex q-PCR, such as Real Time quantitative PGR (qRT- PCR), which enables simultaneous amplification and quantification of at least two miRNAs of interest in one reaction volume by using more than one pair of primers and/or more than one probe. The primer pairs comprise at least one amplification primer that uniquely binds each miRNA, and the probes are labeled such that they are distinguishable from one another, thus allowing simultaneous quantification of multiple miRNAs. Multiplex qRT-PCR has research and diagnostic uses, including but not limited to detection of miRNAs for diagnostic, prognostic, and therapeutic applications.
[0035] The qRT-PCR reaction may further be combined with the reverse transcription reaction by including both a reverse transcriptase and a DN A-based thermostable DNA polymerase. When two polymerases are used, a "hot start" approach may be used to maximize assay performance (U.S. Pat. Nos. 5,41 1,876 and 5,985,619). For example, the components for a reverse transcriptase reaction and a PGR reaction may be sequestered using one or more thermoactivation methods or chemical alteration to improve polymerization efficiency (U.S. Pat. Nos. 5,550,044, 5,413,924, and 6,403,341 ).
[0036] In certain embodiments, labels, dyes, or labeled probes and/or primers are used to detect amplified or unamplified miRNAs. The skilled artisan will recognize which detection methods are appropriate based on the sensitivity of the detection method and the abundance of the target. Depending on the sensitivity of the detection method and the abundance of the target, amplification may or may not be required prior to detection. One skilled in the art will recognize the detection methods where miRNA amplification is preferred.
[0037] A probe or primer may include Watson-Crick bases or modified bases. Modified bases include, but are not limited to, the AEGIS bases (from Eragen Biosciences), which have been described, e.g., in U.S. Pat. Nos. 5,432,272, 5,965,364, and 6,001,983. In certain aspects, bases are joined by a natural phosphodiester bond or a different chemical linkage. Different chemical linkages include, but are not limited to, a peptide bond or a Locked Nucleic Acid (LISA) linkage, which is described, e.g., in U.S. Pat, No. 7,060,809.
[0038] in a further aspect, oligonucleotide probes or primers present in an amplification reaction are suitable for monitoring the amount of amplification product produced as a function of time. In certain aspects, probes having different single stranded versus double stranded character are used to detect the nucleic acid. Probes include, but are not limited to, the 5'-exonuclease assay (e.g., TaqMan™) probes (see U.S. Pat. No. 5,538,848), stem-loop molecular beacons (see, e.g., U.S. Pat. Nos. 6, 103,476 and 5,925,517), stemfess or lineal- beacons (see, e.g., WO 9921881, U.S. Pat. Nos. 6,485,901 and 6,649,349), peptide nucleic acid (PNA) Molecular Beacons (see, e.g., U.S. Pat. Nos. 6,355,421 and 6,593,091 ), linear PNA beacons (see, e.g. U.S. Pat. No. 6,329,144), non-FRET probes (see, e.g., U.S. Pat. No. 6, 150,097), Sunrise™/AmplifiuorB™probes (see, e.g., U.S. Pat. No. 6,548,250), stem-loop and duplex Scorpion™ probes (see, e.g., U.S. Pat. No, 6,589,743), bulge loop probes (see, e.g., U.S. Pat. No. 6,590,091 ), pseudo knot probes (see, e.g., U.S. Pat. No. 6,548,250), cyclicons (see, e.g., U.S. Pat, No. 6,383,752), MGB Eclipse™ probe (Epoch Biosciences), hairpin probes (see, e.g., U.S. Pat. No. 6,596,490), PNA light-up probes, antiprimer quench probes (Li et al, Clin. Chem. 53:624-633 (2006)), self-assembled nanoparticie probes, and ferrocene-modified probes described, for example, in U.S. Pat. No. 6,485,901.
[0039] In certain embodiments, one or more of the primers in an amplification reaction can include a label In yet further embodiments, different probes or primers comprise detectable labels that are distinguishable from one another, in some embodiments a nucleic acid, such as the probe or primer, may be labeled with two or more distinguishable labels.
[0040] in some aspects, a label is attached to one or more probes and has one or more of the following properties: (i) provides a detectable signal; (ii) interacts with a second label to modify the detectable signal provided by the second label, e.g., FRET (Fluorescent
Resonance Energy Transfer); (iii) stabilizes hybridization, e.g., duplex formation; and (iv) provides a member of a binding complex or affinity set, e.g., affinity, antibody-antigen, ionic complexes, hapten-ligand (e.g., biotin-avidin). In still other aspects, use of labels can be accomplished using any one of a large number of known techniques employing known labels, linkages, linking groups, reagents, reaction conditions, and analysis and purification methods.
[0041] MiRNAs can be deiected by direct or indirect methods, in a direct detection method, one or more miRNAs are detected by a detectable label that is linked to a nucleic acid molecule. In such methods, the miRNAs may be labeled prior to binding to the probe.
Therefore, binding is detected by screening for the labeled miRNAs that is bound to the probe. The probe is optionally linked to a bead in the reaction volume.
[0042] In certain embodiments, nucleic acids are detected by direct binding with a labeled probe, and the probe is subsequently detected. In one embodiment of the present invention, the nucleic acids, such as amplified miRNAs, are detected using FlexMAP Microspheres (Luminex) conjugated with probes to capture the desired nucleic acids.
[0043] Some methods may involve detection with polynucleotide probes modified with fluorescent labels or branched DNA (bDNA) detection, for example.
[0044] In other embodiments, nucleic acids are detected by indirect detection methods. For example, a biotinylated probe may be combined with a streptavidin-conjugated dye to detect the bound nucleic acid. The streptavidin molecule binds a biotin label on amplified miRNAs, and the bound miRNA is detected by detecting the dye molecule attached to the streptavidin molecule. In one embodiment, the streptavidin-conjugated dye molecule comprises
Phycolink® Streptavidin R-Phycoerythrm (PROzyme). Other conjugated dye molecules are known to persons skilled in the art.
[0045] Labels include, but are not limited to: light-emitting, light-scattering, and light- absorbing compounds which generate or quench a detectable fluorescent, chemiiuminescent, or bioluminescent signal (see, e.g., Kricka, L., Nonisotopic DNA Probe Techniquies, Academic Press, San Diego (1992) and Ganman A ., on-Radioactive Labeling, Academic Press (1997). Fluorescent reporter dyes useful as labels include, but are not limited to, fluoresceins (see, e.g., U.S. Pat. Nos. 5, 188,934, 6,008,379, and 6,020,481), rhodamines (see, e.g., U.S. Pat. Nos. 5,366,860, 5,847, 162, 5,936,087, 6,051,719, and 6, 191 ,278),
benzophenoxazincs (see, e.g., U.S. Pat. No. 6, 140,500), energy-transfer fluorescent dyes, comprising pairs of donors and acceptors (see, e.g., U.S. Pat. Nos, 5,863,727; 5,800,996; and 5,945,526), and cyanines (see, e.g., WO 9745539), iissamine, phycoerythrin, Cy2, Cy3, Cy3.5, Cy5, Cy5.5, Cy7, FluorX (Amersham), Alexa 350, Alexa 430, AMCA, BOD1PY 630/650, BODTPY 650/665, BODIPY-FL, BODIPY-R6G, BODIPY-TMR, BODIPY-TRX, Cascade Blue, Cy3, Cy5, 6-FAM, Fluorescein Isothiocyaoate, HEX, 6-JOE, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, REG, Rliodamine Green, Rhodamine Red, Renographin, ROX, SYPRO, TAMRA, Tetramethylrhodamine, and/or Texas Red, as well as any other fluorescent moiety capable of generating a detectable signal. Examples of fluorescein dyes include, but are not limited to, 6-carboxyfluorescein, 2',4',1 ,4,- teirachlorofluorescein and 2',4',5',7', 1 ,4-hexachlorofluorescein. In certain aspects, the fluorescent label is selected from SYBR-Green, 6-carboxyfluorescein ("FAM"), TET, ROX, VICTM, and JOE. For example, in certain embodiments, labels are different fluorophores capable of emitting light at different, spectrally-resolvable wavelengths (e.g., 4-differently colored fluorophores); certain such labeled probes are known in the art and described above, and in U.S. Pat. No. 6, 140,054. A dual labeled fluorescent probe that includes a reporter fluorophore and a quencher fluorophore is used in some embodiments. It will be appreciated that pairs of fluorophores are chosen that have distinct emission spectra so that they can be easily dis inguished.
[0046] In still a further aspect, labels are hybridization-stabilizing moieties which serve to enhance, stabilize, or influence hybridization of duplexes, e.g., intercalators and intercalating dyes (including, but not limited to, ethidium bromide and SYBR-Green), minor-groove binders, and cross-linking functional groups (see, e.g., Blackburn et ak, eds. "DNA and RNA Structure" in Nucleic Acids in Chemistry and Biology (1996)).
[0047] In further aspects, methods relying on hybridization and/or ligation to quantify miRNAs may be used, including oligonucleotide ligation (OLA) methods and methods that allow a distinguishable probe that hybridizes to the target nucleic acid sequence to be separated from an unbound probe. As an example, HARP-like probes, as disclosed in U.S. Publication No, 2006/0078894 may be used to measure the amount of miRNAs. In such methods, after hybridization between a probe and the targeted nucleic acid, the probe is modified to distinguish the hybridized probe from the unhybridized probe. Thereafter, the probe may be amplified and/or detected. In general, a probe inactivation region comprises a subset of nucleotides within the target hybridization region of the probe. To reduce or prevent amplification or detection of a HARP probe that is not hybridized to its target nucleic acid, and thus allow detection of the target nucleic acid, a post-hybridization probe inactivation step is carried out using an agent which is able to distinguish between a HARP probe that is hybridized to its targeted nucleic acid sequence and the corresponding unhybridized HARP probe. The agent is able to inactivate or modify the unhybridized HARP probe such that it cannot be amplified.
[0048] In an additional embodiment of the method, a probe ligation reaction may be used to quantify miRNAs. In a Multiplex Ligation-dependent Probe Amplification (MLPA) technique (Schouten et al, Nucleic Acids Research 30:e57 (2002)), pairs of probes which hybridize immediately adjacent to each other on the target nucleic acid are li gated to each other only in the presence of the target nucleic acid. In some aspects, MLPA probes have flanking PCR primer binding sites. MLPA probes can only be amplified if they have been li gated, thus allowing for detection and quantification of miRNA biomarkers.
[0049] Example
[0050] The following examples illustrate various embodiments of the present inventions and are not intended to limit the scope of the invention.
[0051] Global normalization on the miRNA expression data of samples from subjects with mild TBI (mTBI), severe TBI (sTBI), and orthopedic injury to control samples was performed, and candidates for each group were identified. Human serum samples (n=8 each) were collected from each of subjects with mTBI, sTBI, and orthopedic injury. The samples were collected within 24hr of injury. Control samples (n=8) were also collected from healthy control subjects.
[0052] 100 μΐ of serum/plasma sample was used to isolate total RNA including miRNA from the samples. MS2 bacteriophage RNA was used as carrier to enhance the miRNA recovery. The miRNA amount in total RNA was analyzed using bioanalyzer for the integrity and quantity. Complementary DNA (cDNA) was synthesized by using 3μ1 out of 16 μΐ elute, and cDNA was preamplified using a preamplification kit. MiRNA profiling was performed using Taqman low density array platform for human miRNAs followed by data analysis. RealTime StatMiner® from Integromics® bioinformatics tool was used for the identification of significantly altered miRNA levels in the serum samples.
[0053] The real time PCR results of the samples from the subjects with the mTBI, sTBI and orthopedic injury were normalized to the real time PCR result of the control sample. Our analysis showed that 79, 70 and 58 miRNAs were significantly modulated in serum samples from the subjects with the mTBI, sTBI and orthopedic injury, respectively. The levels of the miRNAs in the samples from the subjects with the mTBI and sTBI were compared to the level of the miRNAs in the sample from the subjects with the orthopedic injury. The results showed up-regulation of 13 and 17 miRNAs in the samples from the subjects with mTBI and sTBI compared to the modulated level of the miRNAs in the sample from the subjects with the orthopedic injury. These 13 unique miRNAs for mTBI and 17 unique miRNAs for sTBI are listed in Tables 1 and 2 along with their normalized fold changes indicating their level of expression.
[0054] Table 1 : Altered miRNAs as biomarkers of mild TBI.
Figure imgf000016_0001
[0055] Table 2: Altered miRNAs as biomarkers of severe TBI.
Figure imgf000016_0002
8 hsa-miR-34a 5 3.713739072 3.23E-04
Figure imgf000017_0001
17 hsa-miR-942 5 1.87E-04
[0056] Among these, miRNA biomarkers as shown in Table 3 were found to be present in the samples from the subjects with mTBI and sTBI, but not in the samples from the subjects with the orthopedic injury. Comparison of these 5 miRNAs with serum miRNA profiles of animal TBI models revealed similar miRNAs between human and animal serum post injury. Table 3 shows the potential biomarker candidates in mild and severe TBI along with their normalized fold changes indicating their level of expression.
[0057] Table 3: MiRNA biomarkers for mild and severe TBI.
Figure imgf000017_0002
[0058] All references cited herein are incorporated herein by reference in their entirety. To the extent publications and patents or patent applications incorporated by reference contradict the disclosure contained in the specification; the specification will supersede any contradictory material.

Claims

What is Claimed is:
1. A method of diagnosing traumatic brain injury (TBI) in a subject, the method
comprising a) determining a level(s) of one or more microRNAs (miRNAs) in a biological sample taken from the subject, and b) comparing the determined level(s) of the one or more miRNAs against a level(s) of the same one or more miRNAs from a control subject determined not to be suffering from TBI, wherein an increase in the level(s) of the one or more miRNAs compared to the level(s) of the one or more miRNAs from the control subject determined not to be suffering from TBI is indicative that the subject may be suffering from TBI.
2. The method of claim 1, wherein the one or more miRNAs is selected from the group consisting of miR-194, miR-361, miR-625*, miR-1255B, miR-381, miR-425*, miR- 638, miR-93, miR-328, miR-151-5p, miR-362-3p, miR-486, miR-942, miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR-130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, and miR-491.
3. The method of claim 1, wherein the one or more miRNAs comprise at least miR-328, miR-362-3p and miR-486.
4. The method of claim 3, wherein the one or more miRNAs comprise at least miR-328, miR-362-3p and miR-486, plus one or more of miR-194, miR-361, miR-625*, miR- 1255B, miR-381, miR-425*, miR-638, miR-93, miR-151-5p, miR-942, miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR-130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, and miR-491.
5. The method of any of claims 1-4, wherein the TBI is mild TBI (mTBI) or severe TBI (sTBI).
6. The method of any of claims 1-5, wherein the TBI is a closed head injury (CHI) or a blast-induced traumatic brain injury (bTBI).
7. The method of any of claims 1-6, wherein the subject is human.
8. The method of any of claims 1-7, wherein the biological sample is blood, serum, plasma, cerebrospinal fluid, urine, saliva or tissue.
9. The method of any of claims 1-8, wherein the biological sample is taken from the subject less than a day after a suspected traumatic episode.
10. The method of any of claims 1-8, wherein the biological sample is taken from the subject more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days after the suspected traumatic episode.
11. The method of any of claims 1-10, wherein the subject is at risk of suffering from TBI.
12. The method of any of claims 1-11, wherein the level(s) of one or more specific
miRNAs are determined by a real time PCR.
13. A method of monitoring the progression of traumatic brain injury (TBI) in a subject, the method comprising a) analyzing at least two biological samples from the subject taken at different time points to determine a level(s) of one or more specific microRNAs (miRNAs) in each of the at least two biological samples, and b) comparing the determined level(s) of the one or more specific miRNAs over time to determine if the subject's level(s) of the one or more specific miRNAs is changing over time, wherein an increase in the level(s) of the one or more specific miRNAs over time is indicative that the subject's risk of suffering from TBI is increasing over time.
14. The method of claim 13, wherein the one or more miRNAs is selected from the group consisting of miR-194, miR-361, miR-625*, miR-1255B, miR-381, miR-425*, miR- 638, miR-93, miR-328, miR-151-5p, miR-362-3p, miR-486, miR-942, miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR-130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, and miR-491.
15. The method of claim 14, wherein the one or more miRNAs comprise at least miR- 328, miR-362-3p and miR-486.
16. The method of claim 15, wherein the one or more miRNAs comprise at least miR- 328, miR-362-3p and miR-486, plus one or more of miR-194, miR-361, miR-625*, miR-1255B, miR-381, miR-425*, miR-638, miR-93, miR-151-5p, miR-942, miR- 1291, miR-19a, miR-601, miR-660, miR-9*, miR-130b, miR-339-3p, miR-34a, miR- 455, miR-579, miR-624, and miR-491.
17. The method of any of claims 13-16, wherein the TBI is mild TBI (mTBI) or severe TBI (sTBI).
18. The method of any of claims 13-17, wherein the TBI is a closed head injury (CHI) or a blast-induced traumatic brain injury (bTBI).
19. The method of any of claims 13-18, wherein the subject is human.
20. The method of any of claims 13-19, wherein the biological sample is blood, serum, plasma, cerebrospinal fluid, urine, saliva or tissue.
21. The method of any of claims 13-20, wherein at least one of the two biological samples from the subject is taken from the subject less than a day after a suspected traumatic episode.
22. The method of any of claims 13-20, wherein at least one of the two biological samples from the subject is taken from the subject more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13 or 14 days after the suspected traumatic episode.
23. The method of any of claims 13-22, wherein the subject is at risk of suffering from TBI.
24. The method of any of claims 13-23, wherein the level(s) of one or more specific
miRNAs are determined by a real time qPCR.
25. A method of detecting a microRNA or plurality of microRNA's in a biological
sample, comprising: obtaining a first biological sample from a subject presenting with clinical symptoms of a traumatic brain injury; contacting said first biological sample with a probe for binding at least one microRNA selected from the group consisting of miR-194, miR-361, miR-625*, miR- 1255B, miR-381, miR-425*, miR-638, miR-93, miR-328, miR-151-5p, miR-362-3p, miR-486, miR-942, miR-1291, miR-19a, miR-601, miR-660, miR-9*, miR-130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, and miR-491, to produce an microRNA-cDNA protein complex, and detecting with Northern blot or a real-time PCR the presence or absence of the microRNA-cDNA complex, wherein the absence of the complex is indicative of the absence of the microRNA in the first biological sample.
26. The method of claim 25, wherein the probe is detectably labeled.
27. The method of any of claims 25-26 wherein said biological sample is blood, serum, plasma, cerebrospinal fluid, urine, saliva or tissue.
28. The method of any of claims 25-27 wherein said biological sample is obtained at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days after the suspected traumatic episode after said subject suffers a traumatic brain injury.
29. The method of any of claims 25-28 further comprising: obtaining a biological sample from said subject at a second time point; contacting said biological sample from said second time point with a probe for binding at least one microRNA selected from the group consisting of miR-194, miR- 361, miR-625*, miR-1255B, miR-381, miR-425*, miR-638, miR-93, miR-328, miR- 151-5p, miR-362-3p, miR-486, miR-942, miR-1291, miR- 19a, miR-601, miR-660, miR-9*, miR-130b, miR-339-3p, miR-34a, miR-455, miR-579, miR-624, and miR- 491, to produce an microRNA-cDNA complex; and detecting with Northern blot or a real-time PCR the presence or absence of the microRNA-cDNA complex in said biological sample from said second time point to track the progression of the traumatic brain injury in the subject.
PCT/US2015/036925 2014-06-20 2015-06-22 Microrna biomarkers for traumatic brain injury and methods of use thereof WO2015196191A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462015383P 2014-06-20 2014-06-20
US62/015,383 2014-06-20

Publications (1)

Publication Number Publication Date
WO2015196191A1 true WO2015196191A1 (en) 2015-12-23

Family

ID=54936180

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/036925 WO2015196191A1 (en) 2014-06-20 2015-06-22 Microrna biomarkers for traumatic brain injury and methods of use thereof

Country Status (1)

Country Link
WO (1) WO2015196191A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017153710A1 (en) * 2016-03-08 2017-09-14 The University Of Birmingham Biomarkers of traumatic brain injury
CN108300788A (en) * 2017-01-13 2018-07-20 中国人民解放军南京军区南京总医院 A kind of micro RNA combination and its application for detecting light-duty brain trauma
JP2018522558A (en) * 2015-07-29 2018-08-16 ザ ヘンリー エム. ジャクソン ファウンデーション フォー ザ アドヴァンスメント オブ ミリタリー メディシン インコーポレイテッド MicroRNA biomarkers for traumatic brain injury and methods of use thereof
RU2771757C2 (en) * 2016-03-08 2022-05-11 Дзе Юниверсити Оф Бирмингем Biomarkers of traumatic brain injury

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130022982A1 (en) * 2009-09-14 2013-01-24 Kevin Ka-Wang Wang Micro-rna, autoantibody and protein markers for diagnosis of neuronal injury
US20130158096A1 (en) * 2010-05-28 2013-06-20 Arie Reijerkerk Mirnas involved in the blood brain barrier function

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130022982A1 (en) * 2009-09-14 2013-01-24 Kevin Ka-Wang Wang Micro-rna, autoantibody and protein markers for diagnosis of neuronal injury
US20130158096A1 (en) * 2010-05-28 2013-06-20 Arie Reijerkerk Mirnas involved in the blood brain barrier function

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2018522558A (en) * 2015-07-29 2018-08-16 ザ ヘンリー エム. ジャクソン ファウンデーション フォー ザ アドヴァンスメント オブ ミリタリー メディシン インコーポレイテッド MicroRNA biomarkers for traumatic brain injury and methods of use thereof
EP3329005A4 (en) * 2015-07-29 2019-05-01 The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Microrna biomarkers for traumatic brain injury and methods of use thereof
EP3712277A1 (en) * 2016-03-08 2020-09-23 The University Of Birmingham Biomarkers of traumatic brain injury
EP3712280A1 (en) * 2016-03-08 2020-09-23 The University of Birmingham Biomarkers of traumatic brain injury
CN109415769A (en) * 2016-03-08 2019-03-01 伯明翰大学 The biomarker of traumatic brain injury
JP2019509044A (en) * 2016-03-08 2019-04-04 ザ ユニバーシティ オブ バーミンガム Biomarkers for traumatic brain injury
EP4257705A3 (en) * 2016-03-08 2024-01-17 The University of Birmingham Biomarkers of traumatic brain injury
CN110291210A (en) * 2016-03-08 2019-09-27 伯明翰大学 The biomarker of traumatic brain injury
US10563262B2 (en) 2016-03-08 2020-02-18 The University Of Birmingham Biomarkers of traumatic brain injury
JP2020511953A (en) * 2016-03-08 2020-04-23 ザ ユニバーシティ オブ バーミンガム Biomarkers of traumatic brain injury
WO2017153710A1 (en) * 2016-03-08 2017-09-14 The University Of Birmingham Biomarkers of traumatic brain injury
WO2018138468A1 (en) * 2016-03-08 2018-08-02 The University Of Birmingham Biomarkers of traumatic brain injury
JP7413455B2 (en) 2016-03-08 2024-01-15 ザ ユニバーシティ オブ バーミンガム Biomarkers of traumatic brain injury
RU2771757C2 (en) * 2016-03-08 2022-05-11 Дзе Юниверсити Оф Бирмингем Biomarkers of traumatic brain injury
JP7111619B2 (en) 2016-03-08 2022-08-02 ザ ユニバーシティ オブ バーミンガム Biomarkers for traumatic brain injury
US11414705B2 (en) 2016-03-08 2022-08-16 The University Of Birmingham Salivary biomarkers of brain injury
JP7132224B2 (en) 2016-03-08 2022-09-06 ザ ユニバーシティ オブ バーミンガム Biomarkers for traumatic brain injury
CN109415769B (en) * 2016-03-08 2022-09-30 伯明翰大学 Biomarkers for traumatic brain injury
EP3574117B1 (en) * 2016-03-08 2023-08-16 The University of Birmingham Biomarkers of traumatic brain injury
AU2017231845B2 (en) * 2016-03-08 2023-09-28 The University Of Birmingham Biomarkers of traumatic brain injury
CN108300788B (en) * 2017-01-13 2021-07-09 中国人民解放军南京军区南京总医院 Micro ribonucleic acid composition for detecting light brain trauma and application thereof
CN108300788A (en) * 2017-01-13 2018-07-20 中国人民解放军南京军区南京总医院 A kind of micro RNA combination and its application for detecting light-duty brain trauma

Similar Documents

Publication Publication Date Title
US7993831B2 (en) Methods of normalization in microRNA detection assays
JP5890686B2 (en) How to detect sepsis
US20210277475A1 (en) MicroRNA Biomarkers for Traumatic Brain Injury and Methods of Use Thereof
AU2012275556B2 (en) MicroRNA biomarkers indicative of Alzheimer&#39;s Disease
WO2017055487A2 (en) A METHOD FOR DIAGNOSING A DISEASE BY DETECTION OF circRNA IN BODILY FLUIDS
EP2971084A1 (en) Methods of detecting cancer
CN107858434B (en) Application of lncRNA in liver cancer diagnosis and prognosis prediction
WO2011163214A2 (en) Microrna profiles for evaluating multiple sclerosis
WO2014114802A1 (en) Non-invasive prenatal genetic diagnostic methods
Wang et al. A customized quantitative PCR microRNA panel provides a technically robust context for studying neurodegenerative disease biomarkers and indicates a high correlation between cerebrospinal fluid and choroid plexus microRNA expression
WO2015196191A1 (en) Microrna biomarkers for traumatic brain injury and methods of use thereof
WO2015171510A2 (en) Circulatory micrornas (mirnas) as biomarkers for diabetic retinopathy (dr) and age-related macular degeneration (amd)
CN104480106A (en) Serum/plasma micro-RNA marker for detecting patients with mild cognitive impairment and application thereof
CN109468373A (en) A kind of biomarker relevant to Parkinson&#39;s occurrence and development
US10358679B2 (en) MicroRNA biomarkers for posttraumatic stress disorder and methods of use thereof
KR102276224B1 (en) Composition for diagnosing nontuberculous mycobacterial infection or infection disease
US20200308647A1 (en) Mirnas as biomarkers for alzheimer&#39;s disease
WO2016177774A1 (en) Method of quantifying mirnas using normalization
KR20240053099A (en) Biomarker composition for early diagnosis of gastric cancer and diagnostic method using the same
OA20986A (en) Salivary biomarkers of brain injury.
CN117417997A (en) Genome biomarker combination for diagnosing keratoconus
AU2013204118A1 (en) MicroRNA biomarkers indicative of Alzheimer&#39;s Disease

Legal Events

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

Ref document number: 15809928

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15809928

Country of ref document: EP

Kind code of ref document: A1