WO2006125188A1 - Methodes et systemes d'analyse d'images neuronales microscopiques tridimensionnelles - Google Patents

Methodes et systemes d'analyse d'images neuronales microscopiques tridimensionnelles Download PDF

Info

Publication number
WO2006125188A1
WO2006125188A1 PCT/US2006/019589 US2006019589W WO2006125188A1 WO 2006125188 A1 WO2006125188 A1 WO 2006125188A1 US 2006019589 W US2006019589 W US 2006019589W WO 2006125188 A1 WO2006125188 A1 WO 2006125188A1
Authority
WO
WIPO (PCT)
Prior art keywords
spine
image
dendrite
neuron
program code
Prior art date
Application number
PCT/US2006/019589
Other languages
English (en)
Inventor
Stephen T. C. Wong
Xiaoyin Chen
Xiaoyin Xu
Original Assignee
The Brigham And Women's Hospital, 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 The Brigham And Women's Hospital, Inc filed Critical The Brigham And Women's Hospital, Inc
Publication of WO2006125188A1 publication Critical patent/WO2006125188A1/fr

Links

Classifications

    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06VIMAGE OR VIDEO RECOGNITION OR UNDERSTANDING
    • G06V20/00Scenes; Scene-specific elements
    • G06V20/60Type of objects
    • G06V20/69Microscopic objects, e.g. biological cells or cellular parts

Definitions

  • Neurons are the basic information processing structures in the central nervous system. Each neuron forms thousands of connections with other neurons via axonal and dendritic arborizations. Such connections, known as synapses, are crucial to the biological computations that underlie perception and thought, and provide the means through which the nervous system connects to and controls the other systems of the body (e.g., muscles, glands, etc .).
  • the dendritic tree represents the most substantial receptive area of the nerve cells.
  • Dendritic arborizations receive and integrate incoming synaptic potentials with complex spatio-temporal patterning, and trigger the neuron to generate and transmit time-structured potentials via their axonal arborizations to local and remote target neurons.
  • Dendritic spines are morphological specializations that protrude from the main shaft of neuron dendrites. They play an important role in synaptic transmission by creating a local synapse-specific compartment (G.M. Shepherd, J. NeurophysioL, 1996, 75: 2197-2210). Typically 0.5 to 2 microns in length, dendritic spines come in a wide variety of shapes, and are arbitrarily classified on the basis of their structure as filopodium, thin, stubby, mushroom and cup shaped as shown on Figure 1 (H. Hering and M. Sheng, Nature, 2001, 2: 880-888). Since modern biophysical techniques have begun to reveal the structure-function relationship of dendritic spines (H. Hering and M.
  • Dendritic spines are also lost or distorted in the brains of patients after seizures and strokes and those suffering from dementia, brain tumors, depression, schizophrenia, and chronic alcoholism (M.E. Scheibel et al, Epilepsia, 1974, 15: 55- 80; J. Spacek, Acta Neuropathol., 1987, 73: 77-85; I. Catala et al, Hum. Neurobiol, 1988, 6: 255-259; I.
  • the present invention relates to a new, powerful class of informatics tools for efficient neuron images studies. More specifically, improved systems and strategies are described herein that can be used to automatically or semi-automatically process and analyze neuronal image stacks (i.e., 3D images) obtained by optical microscopy.
  • the present invention provides processes and apparatus with increased capacity to detect and extract neuron components, including axonal arborizations, dendrites, and dendritic spines, and to calculate biologically significant features of dendrites and spines, such as number, density, length, volume, cross-section, surface area, shape, and the like.
  • the inventive systems reduce human bias to the minimum.
  • the present invention provides a method for characterizing one or more neurons, wherein each neuron comprises one or more dendrites and each dendrite comprises one or more spines.
  • the inventive method comprises steps of: receiving a neuron image showing at least one neuron or part of one neuron; performing a segmentation analysis of the neuron image to obtain a segmented digital image; and extracting one or more parameters from the segmented digital image to characterize the at least one neuron.
  • the neuron image is a 3D neuron image, for example, a 3D neuron image obtained using confocal laser scan microscopy or multi-photon laser scan microscopy.
  • the inventive method further comprises steps of: processing the neuron image using a median filter to remove noises and obtain a noise-free neuron image; and deconvoluting the noise-free neuron image to restore image distortion to obtain a processed neuron image prior to performing the segmentation analysis.
  • the median filter may be a center-weighted median filter.
  • the step of performing a segmentation analysis may comprises: applying an unsharp masking filter to the neuron image to obtain a local contrast image; and applying a global threshold process using a preset brightness level to the local contrast image to separate dendrite from background.
  • the segmented digital image obtained comprises a representation of the at least one neuron, each representation comprising a collection of signal intensity values at positions in the image where the neuron is present.
  • the inventive method further comprises a step of applying a high level threshold process to the local contrast image to extract dendrite backbone.
  • applying a high threshold process may be performed by using a medial-axis transformation.
  • the inventive method further comprise removing false positive signals from the local contrast image.
  • This can be achieved by: applying a low level threshold process to the local contrast image to obtain a low level processed image showing dendrites, detached objects, attached objects and smears; measuring, for each detached object, the distance between the detached object and the nearest dendrite backbone; and identifying the detached object as a false positive signal if the distance measured is higher than a present distance value D; and measuring, for each attached object of intensity below the preset brightness level used in the global threshold process, the distance between the attached object and the nearest dendrite backbone in the z-direction; and identifying the attached object as a false positive signal if the distance measured is larger than 2 layers; thereby obtaining a processed image showing dendrites, detached objects and attached objects but no false positive signals except for smears.
  • the preset distance value D may be chosen to be higher than the length of the longest spine.
  • the inventive method further comprises a step of detecting spines in the processed image with no false positive signals except for smears.
  • Detecting spines may comprise steps of: for each detaches object, measuring the length Li of the detached object in the direction parallel to the nearest dendrite backbone; measuring the length L 2 of the detached object in the direction perpendicular to the nearest dendrite backbone; and identifying the detached object as a smear if Li> ⁇ 2 and as a spine head if Li ⁇ 2; and for each object attached to a dendrite, applying a grassfire process from dendrite backbone to dendrite surface to determine voxels of the attached object assigned the locally largest distances by the grassfire process and identifying these voxels as spine base tip; applying a reverse grassfire process from spine base tip to dendrite backbone to obtain sets of voxels of the attached object that are assigned an identical integer by the reverse grassfire process; determining, among the sets of voxels, the set of voxels, the set of
  • the inventive method further comprises a step of merging spine head and spine base that belong to the same spine.
  • the step of merging spine head and spine base that belong to the same spine comprises steps of: calculating the center of mass, Ri, for each spine head; calculating the center of mass, R 2 , for each spine base comprising a spine base tip and a ring of spine-surface boundaries points, measuring the distance between the center of mass Ri and a spine head and the center of mass R 2 of a spine base; and merging the spine head and spine base if the distance measured is lower than a present distance value D ' and the tip of the spine base lies within a cone determined by the center of mass of the spine head and the ring of spine-surface boundaries points.
  • the step of extracting one or more parameters from the segmented digital image to characterize at least one neuron comprises determining one or more features selected from the group consisting of dendrite number, dendrite density, dendrite length, dendrite volume, dendrite shape, dendrite location, spine number, spine density, spine location, spine length (l s ), spine volume, spine surface area, spine head diameter (d / i), spine head width, spine neck diameter (d n ), and spine neck width.
  • the inventive method further comprises classifying at least one spine of a dendrite as f ⁇ lopodium, thin, stubby, mushroom-shaped or cup- shaped.
  • Classifying may comprise calculating ratios of spine length (I 1 ), spine head diameter (d h ), and spine neck diameter (Rvalues for said spine.
  • the one or more neurons in the neuron images may be primary neurons, secondary neurons or immortalized neurons.
  • the neurons are human neurons.
  • the neurons imaged may have been treated under controlled conditions, for example, they may have been treated with a test agent.
  • the present invention also provides machine-readable media on which are provided program instructions for performing one or more of the inventive processes of neuron image analysis.
  • the present invention provides computer products comprising a machine-readable medium on which are provided program instructions for performing one or more of the inventive processes.
  • the present invention provides a neuron image analysis apparatus comprising a memory or buffer adapted to store, at least temporarily, one or more images acquired by optical microscopy, and a processor configured or designed to perform one or more of the inventive processes.
  • Figure 1 shows a scheme illustrating the morphological classification of dendritic spines.
  • Figure 2 is a process flow diagram depicting, at a high level, the system architecture of one embodiment of the inventive method of neuron image analysis.
  • Figure 3 shows two sets of neuron images submitted to image processing according to the present invention.
  • Figure 4 is a process flow diagram depicting, at a high level, the system architecture of one embodiment of the analysis step of the inventive neuron image analysis involving (A) dendrite segmentation and dendrite backbone extraction; (B) false positive signals removal; and (C) spine detection.
  • Figure 5 shows two sets of images illustrating the problems arising when applying global thresholding on neuron images.
  • Figure 6 shows two sets of images illustrating dendrite segmentation based on local contrast map using different threshold values according to the inventive method.
  • A Grey level images
  • B corresponding local contrast maps
  • D) threshold 4;
  • E) threshold 6;
  • F) threshold 8; and
  • G 10
  • Figure 7 shows two sets of images illustrating image segmentation results before and after false positive signals removal.
  • A Images superimposed with dendrite backbones;
  • C binarized image from (B) after false positive signals removal.
  • Figure 8 is a picture showing several examples of detached spine heads and smears.
  • Figure 9 is a scheme showing the segmentation of a theoretical attached spine according to the present invention.
  • Figure 10 presents 4 graphs showing the intersection plane area changes along the center line of the following types of spines: (A) filopodium spine; (B) thin spine; (C) Mushroom spine; and (D) Stubby spine.
  • Figure 11 is a scheme presenting the pipeline of NeuronlQ, one embodiment of the present invention.
  • the pipeline consists of data acquisition from both optical microscopy experiments of neuronal morphology and associated meta-data of genetic information, biological background, and experimental protocols about each image; image pre-processing; image segmentation; extraction of dendrite backbone; spine detection; feature extraction; and statistical analysis.
  • the database includes a relational database and an image repository.
  • Figure 12 shows two images comparing the spine detection results obtained (A) by manual detection and (B) using NeuronlQ detection.
  • Figure 13 is a set of two graphs showing spine length measurements comparison (i.e., as obtained by NeuronlQ and by manual analysis) for (A) apical dendrites and (B) basal dendrites.
  • Figure 14 is an image showing the results of recursive thinning performed using NeuronlQ to extract the backbone of dendrites.
  • Figure 15 is a set of images.
  • A shows a grayscale neuronal image after MIP
  • B shows the segmented image
  • C shows the image obtained using NeuronlQ, wherein spines are marked
  • D shows the same image where spines are detected manually. All the processing is performed in 3D images; it is only for the purpose of illustration that the results are shown in 2D.
  • Figure 16 shows a schematic illustration of a spine length, diameter of the spine head and spine neck.
  • Figure 17 is a set of 4 graphs showing the results of a Kolmogorov-Smirnov test of spine lengths obtained by manual analysis and NeuronlQ from four images stacks (A-D). Blue line, results of NeuronlQ, red line, results of manual analysis.
  • A Manual analysis and NeuronlQ detect 83 and 85 spines respectively
  • B manual analysis and NeuronlQ detect 106 and 104 spines respectively
  • C manual analysis and NeuronlQ detect 59 and 58 spines respectively
  • D manual analysis and NeuronlQ detect 78 and 76 spines respectively.
  • Figure 18 is a graph showing the results of manual analysis and NeuronlQ used to study the effects of TSC2 loss on spine density.
  • Figure 19 presents the tree structure of organizing the neuron image database ofNeuronlQ.
  • the present invention more specifically relates to processes (methods) and apparatus with increased capacity to detect and extract neuron components (including dendrites and dendritic spines) and the ability to identify, analyze and quantitate morphological features of these neuron components from large amounts of images acquired using optical microscopy. Furthermore, based on certain extracted features, the inventive processes and apparatus can classify dendritic spines based on their structure.
  • the present invention also relates to machine-readable media on which are provided program instructions, data structures, etc, for performing one or more of the inventive processes.
  • the inventive image analysis process starts when one or more image analysis tools (typically logic implemented in hardware and/or software) obtain one or more neuron images showing dendrites and dendritic spines of one or more nerve cells.
  • a neuronal image stack i.e., a 3D image is obtained at the beginning of the image analysis process.
  • the image(s) provided at the start of the invention process is/are recorded by an image acquisition system (e.g., a confocal laser scan microscope or multi-photon laser scan microscope).
  • the image acquisition system may be directly coupled with the image analysis tool of the present invention.
  • the image(s) to be processed and analyzed are provided by a remote system unaffiliated with the image acquisition system.
  • the images may be acquired by a remote image analysis tool and stored in a database or other repository until they are ready to be analyzed by the image analysis processes/apparatus of the present invention.
  • Imaged neurons to be analyzed may be of animal (e.g., primate, dog, cat, goat, horse, pig, mouse, rat, rabbit, and the like) or human origin.
  • the neuron images used in the present invention are images of living cells.
  • live cell and living cell are used herein interchangeably. They refer to a cell which is considered living according to standard criteria for that particular type of cell, such as maintenance of normal membrane potential, energy metabolism, or proliferative capability.
  • neuron images are taken from brain slices (i.e., thin sections of living brain tissue that can be maintained in vitro).
  • Methods for the preparation of brain slices are well known in the art and have been used and reported for all kinds of mammal species, including human (see, for example, D. M. Kacobowitz et al, Brain Res. Bull, 1994, 33: 461-463; M.E. Rice, Methods, 1999, 18: 144-149; T. Wang and LS. Kass, Methods MoI. Biol, 1997, 72: 1-14; R.W. Verwer et al., J. Cell MoI. Med., 2002, 6: 429-432).
  • a vibratome or a motorized tissue chopper is used to cut brain slices.
  • Slices can be cut from an isolated portion of the brain (e.g., the hippocampus), or slices can be made of the entire hemibrain.
  • Glutamate antagonists or low-calcium buffer solutions are sometimes employed during slice preparation in order to reduce glutamate-mediated toxicity.
  • brain slices are generally placed in an artificial cerebrospinal fluid in a warm oxygenated chamber until use.
  • cells may be primary cells, secondary cells or immortalized cells (i.e., established cell lines). They may have been prepared by techniques well known in the art (for example, isolation from brain tissue) or purchased from immunological and microbiological commercial resources (for example, from the American Type Culture Collection, Manassas, VA).
  • the images used as the starting point of the inventive methods of analysis are obtained from neurons that have been specifically treated and/or imaged under conditions that contrast markers of cellular components of interest from other cellular components and from the background of the image.
  • the neurons are specifically treated and/or imaged under conditions that contrast dendrites and dendritic spines from other cellular components and/or the background of the image.
  • images may be obtained of neurons that have been treated with a chemical agent that specifically renders visible (or otherwise detectable in a region of the electromagnetic spectrum) the neurons' dendrites and dendritic spines.
  • a chemical agent that specifically renders visible (or otherwise detectable in a region of the electromagnetic spectrum) the neurons' dendrites and dendritic spines.
  • Common examples of such agents are colored dyes or fluorescent compounds that bind directly or indirectly (e.g., via antibodies or other intermediate binding agents) to the dendrites.
  • the nerve cells may have been genetically engineered to express a gene encoding a fluorescent marker, such as the green fluorescent protein, GFP (or any of its derivatives).
  • a fluorescent marker such as the green fluorescent protein, GFP (or any of its derivatives).
  • transgenic expression of GFP within any given cell requires simply placing the GFP coding sequence (or slightly modified versions of the sequence) under the transcriptional control of appropriate regulatory sequences.
  • GFP and its derivatives as well as methods for genetically engineering cells to express these biomolecules are well known in the art.
  • the agent or marker is selected such that it generates a detectable signal whose intensity is related (e.g., proportional) to the amount of cell component to which it is bound. Since the absolute magnitude of signal intensity can vary from image to image due to changes in the cell staining/engineering and/or acquisition procedure and/or apparatus, a correction algorithm may be applied to correct the measured intensities, if desired. Such algorithms can easily be developed based on the known response of the optical system used under a given set of acquisition parameters.
  • the images to be used in the methods and apparatus of the present invention are acquired by fluorescence microscopy techniques such as confocal laser scan microscopy and multi-photon laser scan microscopy. These techniques are particularly useful as they enable visualization deep within both living and fixed cells and tissues and afford the ability to collect sharply defined optical sections from which three-dimensional renderings can be created.
  • Confocal laser scan microscopy and multi-photon laser scan microscopy are similar in many respects. However, there also exist differences between these techniques. In both methods, a beam of laser light is focused into a small point at the focal plane of the specimen to be imaged, for example inside a cell loaded with a probe. A computer-controlled scanning mirror can move or scan this beam in the xy direction at the focal plane. The fluorescence emission created by the point as it is scanned in the focal plane is detected by a photomultiplier tube (PMT), and this detection input is then reconstructed by computer hardware into an image. Among the most important differences between multi-photon and confocal laser scan microscopy is the type of laser utilized in these techniques.
  • Lasers commonly employed in confocal microscopy are high-intensity monochromatic light sources such as argon-ion lasers, krypton-ion lasers or air-cooled lasers using argon-krypton mixtures.
  • the basic key to the confocal approach is the use of spatial filtering techniques ⁇ e.g., involving a pin hole placed between the detector and the specimen being imaged) to eliminate out-of-focus light and glare in specimens whose thickness exceeds the immediate plane of focus. This combination leads to the creation of a sharp image or optic section of the situation inside the specimen.
  • solid state pulsed lasers are used (e.g., titanium sapphire lasers).
  • Such lasers emit light off at a longer wavelength than the gas lasers used in confocal microscopy.
  • the energy of the light is lower and absorption of more than one photon is required for the probe to be promoted to an excited state (e.g., in two-photon laser scan microscopy, excitation of the probe to an excited state requires absorption of two photons). Since the probability of a multi-photon event is extremely low and occurs only where the laser light is the most intense (i.e., at the focal point), essentially all the fluorescent light comes from the focal point of interest and no pin hole is required.
  • Confocal laser scan microscopy has the advantage that the lasers used in this technique are considerably less expensive and much easier to operate than the light sources employed in multi-photon laser scan microscopy.
  • the light of longer wavelength (i.e., of lower energy) used in multi-photon microscopy is inherently less damaging to biological material and penetrates deeper into the sample than light of shorter wavelength used in confocal microscopy (S. L. Murov et ah, "Handbook of Photochemistry” , 2 nd Ed., 1993, Marcel Dekker; K.C. Smith (Ed.), "The Science of Photobiology” , 2 nd Ed., 1989, Plenum).
  • multi-photon microscopy is more sensitive than confocal microscopy because all the fluorescence light generated to make an image is sent directly to the detector.
  • Selecting an appropriate imaging technique to acquire neuron images to be analyzed by the processes and apparatus of the present invention can easily be performed by one skilled in the art and will depend on several factors including, but not limited to, the nature of the specimen to be imaged (e.g., brain slice vs. cells in culture dish).
  • the image analysis tool of the present invention performs an image processing step.
  • Image processing aims at producing clean and clear output images that are ideally noise-free and have little or no artifacts. Any method that leads to clean and clear output images may be used in the practice of the present invention to process neuron images.
  • the neuron images are processed by using first a median filter for removing noises, and then a deconvolution processing to restore image distortion.
  • the raw image i.e., as obtained by the image acquisition system
  • the raw image can be considered as a blurred version of the real image plus noise. More specifically, considering the physics of imaging, the observed microscopy image can be modeled as follows:
  • Equation (1) can be written as:
  • noise generally results from the imaging mechanism of the photomultiplier tube (PMT) used as detector.
  • PMT photomultiplier tube
  • a PMT is a high gain, low sensitivity device which, combined with the low number of photons emitted by small structures such as dendritic spines, produces "photon shot noise”.
  • a non-linear filter such as a median filter is generally applied (J.W. Tukey, "Exploratory Data Analysis ' ", 1971, Addison- Wesley: Reading, MA).
  • Standard median filter works by scanning an image using a filter window (typically 3 x 3, 5 x 5 or 7 x 7 pixels in size). In median filtering, each pixel is determined by the median value of pixels in the filter window.
  • center-weighted median filter (CWMF) is preferably used to remove photon shot noise.
  • the filter window W used to scan the 3D image is defined in terms of the image coordinates in the 3D neighborhood centered at the current voxel. For example, a (2L+1) x (2Z+1) x (2Z+1) cubic window is given by:
  • CWMF center-weighted median filter
  • Figure 3(A) shows the maximum intensity projections of two neuron images
  • Figure 3(B) shows the corresponding images obtained after median filtering process.
  • the median filter effectively removes the noise while keeping the dendrites and spines.
  • a deconvolution process may then be applied to the resulting image to restore image distortion caused by the optical point spread function.
  • Any suitable deconvolution system may be used in the practice of the present invention.
  • the present Applicants used a deconvolution package, AutoDeblur (AutoQuant Image, Inc., Troy, NY).
  • Figure 3(C) shows two images obtained after 5 deconvolution iterations performed on the images presented in Figure 3(B).
  • the objective of image analysis is to obtain quantitative information from an image (or image stack) such as number, sizes and shapes of objects.
  • image analysis classifies and extracts biologically significant features about neurons and neuron components from the studied images. These features include, but are not limited to, dendrites and/or spines length, volume, surface area, density, and the like.
  • Dendrite segmentation and spine detection are critical parts of an automated neuron images analysis system as the quality of dendrite segmentation and spine detection affects the accuracy of the subsequent measurements of dendrites and spines features.
  • FIG. 6 A high level process flow diagram in accordance with one embodiment of the inventive image analysis method is depicted in Figure 4. Each step or module of the inventive image analysis process is described in detail below.
  • Segmentation which transfers a grey-scale image to a black-and-white image, is generally the first step performed by the inventive image analysis process.
  • Nishikawa and coworkers have used a global grey-level threshold in a first step, and then a local adaptive thresholding technique in a second step (R.M. Nishikawa et al, Med. Phys., 1993, 20: 1661-1666).
  • Chan and coworkers have applied a local grey level thresholding technique, where the local threshold is varied using the standard deviation of the surrounding pixel values (H.P. Chan et al, Invest. Radiol., 1990, 25: 1102-1110).
  • Kegelmeyer and coworkers have analyzed six different algorithms, including three algorithms based on grey level thresholding and three algorithms that use local contrast estimation (W.P. Kegelmeyer et al, in "Digital Mammography", 1994, Elsevier: Amsterdam, pp. 3-12).
  • the present invention provides an improved segmentation method particularly suited to the segmentation of neuron images.
  • the inventive segmentation method starts by applying an unsharp masking technique to obtain the local contrast images and then a global thresholding technique is performed to separate neurons from their background.
  • Unsharp masking techniques are known in the art and have originally been used in the photographic darkroom. Digital unsharp masking filters stimulate the effect of the true photographic technique, which creates a blurred version of the image and then subtracts it from the original to create a sharper, more detailed image.
  • unsharp masking technique A.R. Cowen et al, SPIE Image Processing, 1993, 1898: 833-843
  • WJ. Vedkamp and N. Karssemeijer IEEE Trans. Med. Imaging, 200, 19: 731-738
  • S. Dippel et al IEEE Trans. Med. Imaging, 2002, 21: 343-353.
  • the present invention extends the use of this method to process and analyze neuron 3D images.
  • the following equation is employed to calculate local contrast C 1 at site i:
  • each voxel is normalized by subtracting the result of moving-average filter fromj ⁇ ,-.
  • a global threshold process is then applied to the local contrast image resulting from unsharp masking, in order to separate dendrites from their background. Thresholding is based on simple, well-known concepts.
  • a parameter, called the brightness threshold is chosen and applied to each voxel of the image under consideration as follows: (1) if the intensity of the voxel is higher than the brightness threshold, the voxel is considered as belonging to an object (e.g., a dendrite); (2) if the intensity of the voxel is lower than the brightness threshold, the voxel is considered as belonging to the background.
  • Voxels within big dendrites that have low local contrast may be removed by application of a global threshold, as high intensity voxels are considered as dendrite even if their contrast is very small, while low local contrast voxels are usually considered as background. To prevent this, the segmentation results are compared to the deconvoluted grey level image and voxels whose values are above a preset dendrite threshold are included in the resulting image.
  • FIG. 6 shows examples of dendrite segmentation obtained at different threshold levels.
  • the segmentation at threshold 2 contains many false positive signals. At threshold 10, most of these false positive signals are removed.
  • the shape of dendrite does not change too much at all as the threshold level is varied.
  • spines lose some voxels at a higher threshold level, they still contain enough voxels to be identified as spines.
  • the segmentation process of the present invention uses a low threshold level to keep most of the spine voxels in the image, and a separate routine is provided herein to remove false positive signals.
  • False positive signals come from different sources such as disconnected dendrites, axons, and smears caused by the optical point spread function. Disconnected dendrites and axons have the same intensity and contrast as the dendrite being analyzed. Fortunately, they are generally separated in space from the dendrite of interest. Thus, disconnected dendrites and axons can be removed by comparing their distance to the dendrite of interest. Most of the smears appear along the optical axis at different layers from the dendrite. They can be removed by comparing their distance to the dendrite of interest in the z direction. Since the position of the dendrite is critical in removing false positive signals, the dendrite backbone is first extracted.
  • dendrite backbones are extracted by applying a high threshold to the local contrast image (see Figure 7(A)).
  • a medial axis algorithm T.C. Lee et al, CVGIP: Graph Models Image Proc, 1994, 56: 462-478
  • a medial-axis transformation is generally useful in thinning a polygon, or, in other words, finding its skeleton.
  • the medial axis transform of a neuron image contains the skeleton of all dendrites and big spines, including the skeleton of disconnected dendrites and spurs.
  • short medial axes are removed and spurs are trimmed.
  • Figure 7(A) shows examples of dendrite backbones superimposed with the corresponding binary dendrite images.
  • a preset value may be selected such that it corresponds to a distance at which no dendritic spine can be found protruding out of a dendrite shaft.
  • a preset value is preferably chosen to be > 2 microns.
  • a preset value of 5 microns was used as no spines can protrude out of the dendritic surface more than 5 microns.
  • Attached objects are separated according to their intensity level. If the intensity level of a dendrite voxel is below the dendrite intensity threshold (i.e., the brightness level used in the global thresholding technique described above), its distance to the dendrite backbone is measured in the z-direction, and it is considered as a false positive signal if this distance is larger than 2 layers.
  • the dendrite intensity threshold i.e., the brightness level used in the global thresholding technique described above
  • Figure 7(C) shows examples of neuron images after removal of false positive signals. After these correction processes have been performed, the only false-positive signals remaining in the image under consideration are smears located within the layer nearby the dendrite backbone. As detailed below, in the analysis process of the present invention these smears are removed during spine detection.
  • the main difficulty of spine segmentation comes from the shape variation of different spine types as well as dendrite and spine surface irregularities.
  • Mushroom spines are characterized by big heads and thin necks. Their necks are generally too thin to be detected by light microscopy. In the binary dendrite images, they appear as detached head-base pairs.
  • Stubby spines on the other hand, have very thick necks. Small stubby spines may only protrude out of the dendrite surface by 0.2 microns, which makes them look very similar to dendrite surface irregularities.
  • Quantification of spine parameters such as length, volume, head width, and neck width requires an accurate segmentation between spine and dendrites.
  • the ideal segmentation plane is the dendrite surface. However, it is generally very difficult to estimate the dendrite width locally due to dendrite and spine surface irregularities.
  • Imaging., 1997, 16: 28-40 have applied multi-level threshold and then analyzed the contours pattern to determine the best segmentation point between objects and their background in medical images.
  • the present invention provides a similar method to detect the transition point from spine to dendrite in order to obtain an accurate segmentation between them.
  • spine detection according to the inventive process allows several spine features to be measured.
  • spine voxels belong to two types of spine components: detached spine components and attached spine components.
  • the present invention provides different methods to detect and identify each type of spine component (see Fig. 4(C)). After detection and identification, a post-processing step is used to merge those spine components that belong to the same spine.
  • the shape of a detached spine candidate is analyzed and classified based on this observation.
  • a detached spine candidate is identified as a smear if its length in the direction parallel to the dendrite backbone is longer ⁇ e.g., 1.2 times longer) than its length in the direction perpendicular to the dendrite backbone.
  • the detached spine candidate is identified as a detached spine component if its length in the direction perpendicular to the dendrite backbone is longer than its length in the direction parallel to the dendrite backbone.
  • a new method is provided herein to detect the transition from spine to dendrite.
  • each voxel in the dendrite is first labeled with an integer time step using a grassfire technique (R. Leymarie and M.D. Kevine, IEEE Trans. Pattern Anal. Mach. Intell, 1992, 14: 56- 75).
  • a grassfire technique the object is imaged to be filled with dry grass and an initial firefront is ignited at the object boundary. It then propagates along the inward normal at constant speed. The time of arrival of the grassfire front at a given point equals the distance of that point from the shape boundary.
  • the shape skeleton is the locus of points where fronts from two or more directions meet.
  • Figure 10 shows the variation of the number of voxels in the intersection planes as a function of time integer step for 4 different morphological types of spines, i.e., filopodium spine, thin spine, mushroom spine and stubby spine.
  • This figure clearly demonstrates that, for each type of spine, the number of voxels in the intersection plane (or intersection area) is the largest when the plane reaches the dendrite surface.
  • the maximum voxel number criterion is used to localize the segmentation plane. However, using this criterion may result in some dendrite voxels being counted as spine voxels.
  • the segmentation plane localized by this method may go deep into the dendrite.
  • the process of the present invention finds the voxel within this plane whose distance to the dendrite backbone is the largest (e.g., distance d shown on Figure 9), and then identifies the segmentation plane between the spine and dendrite as the plane passing through this voxel, and parallel to the dendrite backbone (see Figure 9).
  • the number of voxels within the spine component is then determined and the distance (L) from the spine tip to the segmentation plane is calculated.
  • spine components with less than 10 voxels or whose length was less than 3 microns were considered as false protrusions and were rejected.
  • a detached spine head and an attached spine base must be such that the distance between their centers-of-mass is less than a preset value and the tips of spine base must lie within the cone determined by the center of the spine head and the ring of spine-surface boundaries points.
  • the preset value is preferably selected to be higher than the length of the longest dendritic spines. For example, for 0.5 to 2 micron-long dendritic spines, the preset value may be selected to be 3 microns.
  • Dendrite and Spine Features Measurements. After dendritic segmentation and spine detection, several dendrite and spine parameters, including, but not limited to, spine density, dendrite length and volume, spine length, volume and surface area, spine head width and neck width, can be determined. Any suitable method can be used to quantify these features.
  • a spine length may be determined as follows. For a fully attached spine, the spine length may be determined by subtracted the integer time step of the spine tip from the voxel with the smallest integer time step within the spine. For partially attached spines (consisting of a base and a detached head), the spine length is the sum of base component length and detached component length. The base component length may be measured the same way as fully attached spines. The detached component length is determined as the distance from the base component tip to the farthest spine voxel within the detached component. For a detached spine, a line may be drawn between the center of mass of spine to its closest dendrite backbone. The detached spine length is determined as the distance from the dendrite surface voxel on this line to the farthest spine voxel within the spine.
  • spine lengths can be measured in both 2D and 3D.
  • Manual analysis can only measure spine lengths in 2D by projecting the 3D stack of image slices along the optical direction. So in order to be able to compare the manual and automatic methods, spine lengths in 2D should be calculated (see Examples).
  • Volumes of spines may be calculated by multiplying the number of voxels in the spine with the unit volume of a voxel. The unit volume of a voxel is computed as the product of resolution in the x, y and z directions, taking into account that usually resolution in the z-direction is different from that in the x- and ⁇ -directions.
  • the surface area of a spine may be calculated in a similar manner, by adding the unit area of outward facing side(s) of boundary voxels.
  • Spines shapes may be classified by calculating the ratios between the spine length I s , head diameter dj,, and neck diameter d n .
  • the image analysis methods of the present invention employ various processes involving data stored in or transferred through one or more computer systems. Accordingly, embodiments of the present invention also relate to an apparatus for performing these operations.
  • This apparatus may be specifically constructed for the required purposes, or it may be a general-purpose computer selectively activated or reconfigured by a computer program and/or data structure stored in the computer.
  • the image analysis processes disclosed herein are not inherently related to any particular computer or other apparatus. Actually, the methods of the present invention may be implemented on various general or specific purpose computing systems. In certain embodiments, the image analysis methods of the present invention may be implemented on a specifically configured personal computer or workstation. In other embodiments, the image analysis methods of the present invention may be implemented on a general-purpose network host machine such as a personal computer or workstation. Alternatively or additionally, the methods of the invention may, at least partially, be implemented on a card for a network device or a general-purpose computing device.
  • certain embodiments of the present invention relate to computer readable media or computer program products that include program instructions and/or data (including data structures) for performing various computer implemented operations.
  • Examples of computer readable media include, but are not limited to, magnetic media such as hard disks, floppy disks, and magnetic tapes; optical media such as CD-ROM disks; magneto-optical media; semiconductor memory devices, and hardware devices that are specifically configured to store and perform program instructions, such as read-only memory devices (ROM) and random access memory (RAM).
  • ROM read-only memory devices
  • RAM random access memory
  • the data and program instructions of the present invention may also be embodied on a carrier wave or other transport medium. Examples of program instructions include both machine code, such as produced by a compiler, and files containing higher level code that may be executed by the computer using an interpreter.
  • the present invention provides a neuroinformatics system, featuring software tools with persistent database in an integrated data processing pipeline, for segmentation, quantitation, correlation, and analysis of microscopic neuronal images.
  • One embodiment of the inventive pipeline (called NeuronlQ, i.e., Neuron Image Quantitator) is comprised of five subsystems, namely data acquisition, image processing, image analysis, data analysis, and database, as shown on Figure 11.
  • NeuronlQ processes and analyzes high-resolution neuronal and dendritic images acquired by optical microscopy, extracts features from images, and deposits extracted features and other information into a relational database system for subsequent browsing, exploration, retrieval, and statistical analysis (for example, on the Internet).
  • Image features of interest include soma cross-section area, number of primary dendrites, branch number and length of dendrites, dendritic spine density, length, types, shapes, and their changes over time. Based on the results of image analysis, statistical analysis can be performed to investigate relationship between dendrite and spine morphology and experimental conditions (i.e., a given neural disease or condition of the subject from which neurons have been imaged).
  • NeuronlQ is built on modules for easy maintenance and upgrade.
  • the data acquisition, image processing and image analysis subsystems are implemented in C/C++.
  • the data acquisition relies in identification of regions of interest by the user.
  • the automation effort of Neuron IQ is on the post-processing and management of acquired images (i.e., image processing, analysis, and archival).
  • Data analysis comprises the use of independent statistical packages such as SAS (Statistical Analysis System, http://www.sas.com) and SPSS (http://www.spss.com).
  • the application and web servers of NeuronlQ are based on Appache (http://www.apache.org).
  • the database system is implemented using a structured query language (SQL) relational database. All subsystems work under Linux or Microsoft Windows.
  • the neuronal morphometric features that can be extracted by NeuronlQ are as follows: for the primary dendrites: number and cross-section volume of soma; for the dendrites: number of branches and length of branches; for the dendritic spines: density, length, volume, shape and location.
  • the NeuronlQ data model has three parts, i.e., binary image data, meta-data definitions from image analysis, and associated meta-data definitions of the images, e.g., relevant genetics, biological, and experimental information.
  • the data model is instantiated via a relational database in which meta-data are stored in tables as specified by the schema, and both raw and pre-processed binary image data are stored in an image repository.
  • the image repository is indexed by the pointers stored in the database tables.
  • the NeuronlQ database contains other biological and experimental parameters of the specimens. These descriptive parameters include: type of species; genotype information; sex; age of subject at the time of samples (e.g., brain slices) preparation; brain region sliced; culture media used; days in vitro (DIV) when transfected; plasmids transfected; DIV when imaged; other drugs and times of application; type of cell imaged, type of cell region imaged; type and model of optical microscopy scanners; data from image acquisition protocols, data and time of acquisition, information about the experimenter; and other administrative information.
  • types of species e.g., genotype information; sex; age of subject at the time of samples (e.g., brain slices) preparation; brain region sliced; culture media used; days in vitro (DIV) when transfected; plasmids transfected; DIV when imaged; other drugs and times of application; type of cell imaged, type of cell region imaged; type and model of optical microscopy scanners; data from image acquisition
  • NeuronlQ allows the following features to be extracted: number of spines on each dendrite, length of each dendrite, spine density on each dendrite, and other features for each spine such as layer, volume, head volume, focal volume, length, height, head width, neck width, surface and compactness.
  • the parameters first need to be extracted based on the features of spines on each dendrite. Since the features are in different levels, the inventive system performs a z-score based normalization on each feature, using the following equation:
  • Xy is the/' 1 feature of the i th spine
  • I ⁇ i ⁇ m, l ⁇ j ⁇ n, and m is the total number of spines on one dendrite and n is the total number of features of each spine
  • X j and std(x. y ) are the mean and standard deviation of the j th feature.
  • the distribution is considered as a mixture model.
  • a simple case is considered, i.e., the distribution of features is modeled by a Gaussian probability density function. The mean and variance of each feature is then used to represent the feature.
  • the first neuron in apical region was found to have three dendrites, whose lengths were 496, 293, and 155 microns, and which carried 43, 68, and 12 spines, respectively.
  • the second neuron in basal region had six dendrites whose lengths were 477, 315, 301, 98, 44, and 31 microns, carrying 56, 25, 25, 5, 5, and 2 spines, respectively.
  • the third neuron in basal region had one dendrite whose length was 528, and carrying 1 spine.
  • the Pearson linear correlation coefficients were calculated to be 0.9999 between the first and second neurons, 0.9992 between the first and third neurons, and 0.9997 between the second and third neurons, which led to the conclusion that these three studied neurons are similar to each other.
  • NeuronlQ can be extended to include advanced information processing techniques such as Kolmogorov-Smirnov test, T-test, ANOVA, and other information processing techniques such as feature selection and pattern recognition (X. Zhou et ah, J. Biol. Systems, 2004, 12: 371-386)
  • advanced information processing techniques such as Kolmogorov-Smirnov test, T-test, ANOVA, and other information processing techniques such as feature selection and pattern recognition (X. Zhou et ah, J. Biol. Systems, 2004, 12: 371-386)
  • the processes and apparatus of the present invention will find numerous applications as powerful informatics tools which will help better understand how neuron morphology relates to neuronal function, which is crucial to the development of therapies and drugs for the prevention and/or treatment of neural disorders.
  • the inventive processes and apparatus will provide a reliable, non-bias, automated solution to process and analyze large volumes of microcopy neuron image datasets and to investigate dynamic spine shape changes....
  • the inventive processes and apparatus of neuron image analysis can be used to investigate the dynamic plasticity of dendritic spines. It has been found that over a time course of seconds to minutes, the majority of spines change their shape, and over a matter of hours, a substantial fraction of spines appear or disappear. The rapid morphological changes of spines has raised the possibility that those categories, rather than being intrinsically different populations of spines, represent instead temporal snapshot of a single dynamic phenomenon. The dynamic behavior of spines has attracted particular attention because they are the only neuronal structures that convincingly show experience-dependent morphological changes in the mammalian brain. The methods provided by the present invention may be used to understand these phenomena.
  • Confocal laser scan microscopy (CLSM) and two-photon laser scan microscopy (2PLSM) provide equivalent challenges to image analysis.
  • 2PLSM generally performs better when working with living tissue, which allows high resolution fluorescence imaging of brain slices up to several hundred microns deep with minimal photodamage (B. Lendvai et al, Nature, 2000, 404: 876-881).
  • 2PLSM was used to acquire data.
  • the xy size of the images was 512 x 512 with various z sizes depending on the neuron being studied.
  • the step size of the microscope was 0.07 microns in both x and y direction and 1 micron in the z direction.
  • Ten of the 20 neuron images were from basal area of pyramidal neurons and the other ten were from apical area.
  • Figure 12 gives an example of spine detection result comparison between a human expert and NeuronlQ.
  • the human expert detected a total of 18 spines whereas NeuronlQ detected all those detected by the expert as well as an additional small spine that the human eye has missed.
  • Dendrite segmentation and spine detection are the most important and difficult parts for dendritic spine analysis. The accuracy of the parameters measured depends on the quality of dendrite segmentation and spine detection results.
  • an unsharp mask technique (B. Lendvai et ah, Nature, 2000, 404: 876-881) is used to reduce the high dynamic range of neuron image.
  • a low threshold is then applied to the local contrast map to keep all low contrast dendrite components.
  • a separate procedure is provided to remove the false positive signals caused by disconnected dendrites, axons, and smears.
  • Spine detection in the inventive method is performed after dendrite segmentation. Detached spine components are detected by checking their relative location to dendrite backbone. Attached spine segmentation which separates the spine component from the dendrite component is the most difficult part of spine detection.
  • a new method for spine segmentation is provided herein that comprises employing a grassfire technique to label a possible spine protrusion from its tip to the dendrite. Voxels with the same label in a spine form one intersection plane. In this method, the transition point from spine to dendrite is determined as the place were the number of voxels in the intersection plane increases the most.
  • the voxel, within this intersection plane, which has the largest distance to the dendrite back is identified, and the segmentation plane between the spine and dendrite is determined to be the plane that is parallel to the dendrite backbone and that passes through this voxel.
  • a post-process is then used to merge detached spine heads with attached spines based on their relative location.
  • 3DMA short for 3D Medial Axis, was created by Lindquist et al at SUNY-Stony Brook (CM. Weaver et al, J. Neurosci. Methods, 2003, 124: 197-205; CM. Weaver et al, Neural Comput, 2001, 16: 1353-1383; LY. Y. Koh et al, Neural Comput, 2002, 14: 1283-1310) to analyze neuron images and detect spines.
  • 3DMA is not connected to an associated database and modeling.
  • a method that process grayscale neuronal images was developed by Wearne et al (Neuroscience, 2005, 136: 661-680) using Rayburst sampling algorithm.
  • the Rayburst technique was applied to 3D neuronal shape analysis at different scales to identify spines.
  • NeuronlQ the neuroinformatics system provided herein, features software tools with persistent database in an integrated data processing pipeline for segmentation, quantitation, correlation, and analysis of optical microscopy neuronal images.
  • the pipeline of NeuronlQ consists of five subsystems, data acquisition, image processing, image analysis, data analysis, and database (as shown in Figure 11). Data were populated from neuroscience experiments and obtained from optical microscopy.
  • the automation effort of NeuronlQ currently is in the post-processing and management of the acquired images, i.e., processing, analysis, and archival. It extracts features from images, and deposits the results and other information into a relational database system.
  • the database can be accessed via Internet.
  • Image features of interest include soma cross-section area, number of primary dendrites, branch number and length of dendrites, dendritic spine density, length, types, shapes, and their changes over time.
  • Fluorescence label such as green fluorescence (GFP) was used to mark neurons in vitro. GFP absorbs blue light and converts this light to green light, which is of lower energy. The emitted green light can then be captured by optical microscope like confocal laser scanning microscopy (CLSM) and two-photon laser scanning microscope (2PLSM) and reveals details of the specimen.
  • CLSM confocal laser scanning microscopy
  • 2PLSM two-photon laser scanning microscope
  • Dissociated hippocampal neurons were obtained from mice carrying a conditional Tscl allele (S.F. Tavazoie et al, Nature Neurosci., 2005, 8: 1727-1734). Neurons were transfected by GFP in organotypic hippocampla slices. Three dimensional images were acquired using 2PLSM with an excitation wavelength of 910 nm. The detailed acquisition steps have been described in S.F. Tavazoie et al, Nature Neurosci., 2005, 8: 1727-1734. Images were acquired at 5x magnification at spiny regions of basal and apical dendrites and optical sections were taken at 1.0 ⁇ m spacing. Image Processing and Analysis
  • the recursive thinning process employs a number of templates of size 3 x 3 x 3 centered at each voxel. Each template is rotated and flipped around the central voxel to detect surface voxel and eliminate it from the object.
  • a 6-direction thinning processing is used with 21 templates (K. Palagyi and Q. Kuba, Pattern Recognition Letters, 1998, 19: 613-627).
  • smoothing and trimming is then performed to remove short spurs to obtain the backbone of dendrites (see, for example, Figure 14 obtained using NeuronlQ).
  • spines are detected in the following manner. First, for each detached component, the distance between this component to the nearest spine surface is measured and is used to determine whether this component is an artifact or candidate of spine. Second, for each attached component, two distances are measured. The first distance is measured between the spine candidate protruding out from the dendrite and the dendrite surface. The second distance is measured between the spine candidate protruding into the dendrite and the dendrite surface. The attached component is labeled a spine candidate if it protrudes out farther than it protrudes into the dendrite surface. Third, a merging algorithm checks all the spatial relationships between attached and detached candidates to determine whether some of them are to be merged.
  • NeuronlQ measures volume of spine by multiplying the number of voxels in the spine with the unit volume of a voxel.
  • the unit volume of a voxel is computed as the product of resolution x-, y-, and z-direction (where the resolution in the z-direction is usually different from that of the x-direction and y-direction).
  • NeuronlQ measures the surface area of detected spine in a similar manner, by adding the unit area of outward-facing side(s) of boundary voxels.
  • a spine can be categorized into one of the three types: stubby, thin and mushroom (K. Zito and V.N. Murphy, Current Biology, 2002, 12: R5; LY. Koh et al, Neural Comput, 2002, 14: 1283-1310).
  • Figure 15 shows an example of processed and detected spines. Spine shapes are classified according to the definition given by Harris et al. (J. Neurosci., 1992, 12: 2685-2705). In this method, the spine shape is decided by the length of spine l s , head diameter d h , and neck diameter d n (see Figure 16).
  • NeuronlQ Features measured by NeuronlQ are organized and stored in a neuronal image database for further analysis. Table 3 lists neuronal morphometric features extracted by NeuronlQ. For the purpose of further statistical analysis, the results of NeuronlQ are stored in an associated database as described below.
  • Figure 17 presents the results of Kolmogorov-Smirnov (K-S) test of spine lengths of four different images, which provides another way to compare the cumulative distribution of spin length given by manual analysis and NeuronlQ. There were a total of 367 spines in the four images. From the cumulative distribution plots of Figure 17 (A-D), it can be noted that the two methods generated very similar results in spine length. At a level of 0.05, the K-S tests found no evidence of rejecting the hypotheses that the spine lengths given by manual analysis and NeuronlQ are almost the same.
  • TSC tuberous sclerosis complex
  • TSC is a hamartomatous disorder in which benign tumors proliferate in many organ systems including the brain, heart, kidney, and skin.
  • TSC is caused by lack of the protein product of either TSCl or TSC2 alleles, which form a heterodimer and act as a negative regulator of mammalian target of rapamycin (mTOR), a kinase implicated as a master regulator of cell growth.
  • mTOR mammalian target of rapamycin
  • mTOR mammalian target of rapamycin
  • NeuronlQ was implemented to comprise a neuronal image database to archive neuron images, extracted image features and associated meta-data of the experiments.
  • the image database runs on an Oracle 1Og enterprise server and a 52- CPU Linux cluster, with a link to a digital image repository that has eight terabytes of network attached storage.
  • the NeuronlQ data model has three parts: image data, metadata definitions from image analysis, and associated meta-data definitions of the images, e.g., relevant genetic information, biological background, and experimental protocols.
  • the data model is instantiated via a relational database in which meta-data are stored in tables as specified by the schema, and both raw and pre-processed binary image data are stored in an image repository. Post-processed images are also archived in the database.
  • the image repository is indexed by the pointers stored in the database tables.
  • each microscopy experiments is defined as an imaging session, to which microscopy configurations, extracted image features, and biological and experimental parameters of the specimens are related.
  • the tables of microscopy configurations and equipment are directly associated to imaging session table.
  • Each imaging session consists of several fields of image taken from the same experiment.
  • a field of images represents an image stack of a particular area of tissue with the same configuration of field of view, resolution in three directions, and zoom factor.
  • a field of view may focus on an apical or basal dendrite and this information is stored in the NeuronlQ database.
  • Extracted image features are stored in entities of dendrites and spines.
  • the tree structure of the NeuronlQ database for processed datasets is shown in Figure 19.
  • Features of dendrite include branches, tree number, length, surface area, volume, base diameter, average diameter, base coordinate, density of spines, and tortuosity.
  • Features of spines include length, volume, position, surface area, head width, and neck width (see Table 6). Information about the experimenter and other administration information are stored in the tables of project, organization and staff.
  • Biology and experiment information such as animal and tissue, are stored in animal or tissue tables. These descriptive parameters include: type of species; genotype information; sex; age of subject at preparation of slices; brain region sliced; culture media used; days in vitro (DIV) where transfected; plasmid(s) transfected; DIV when imaged; other drugs and times of application; types of cells imaged; type of cell regions imaged; type and model of optical microscopy scanners; data from image acquisition protocols; data and time of acquisition.
  • DIV days in vitro
  • the first version of a web application was developed by the Applicants on JBoss, an open source J2EE application server.
  • "Apache Torque" a database object persistence code generator, was used to generate Java database access code from an XML database schema definition file. This code generator was chosen because it significantly shortened the development cycle and gave great flexibility in schema change. Apache Torque also can generate database access code for a variety of databases. Porting the web application to support a database other then the current choice of Oracle requires a simple recompilation. Other core technologies used for the web application include Java Servlet, JSP (Java Server Pages), JSTL (Java Standard Tag Library) and JDBC (Java Database Connection). The web application currently supports a simple search on project data by a researcher's name, project keyword and subject species. It also supports experimental data download in CSV (comma-separated values) format.
  • NeuronlQ which is provided herein, is, to our knowledge, the first integrated neuroinformatics systems that allows for the study of dendritic spine morphology. NeuronlQ can be used to analyze perturbations of neuronal morphology caused by genetic mutations related to neurological diseases. For example, NeuronlQ has been successfully used herein to investigate abnormal dendritic spines observed in TSC using a cell-autonomous model.
  • NeuronlQ is able to provide rich collection of quantitative features about neurons, such as length of dendrites, number and density of spines on a dendrite, and shape of a spine, under various experimental conditions. Such features are then incorporated in a persistent database for subsequent data analysis. Data analysis allows comparative studies to be performed not only within an experiment, but among experiments under different settings.
  • NeuronlQ The main advantages of NeuronlQ include: (1) it extracts reproducible and objective feature measurements from neuronal images, and (2) it provides a mechanism for managing and analyzing large amounts of image datasets generated in high resolution optical microscopy by integrating its image processing capability with data analysis and persistent database management. NeuronlQ and associated databases of various neuroscience applications are still evolving. NeuronlQ is one of the first informatics systems dedicated to solve neuronal image management and analysis problems. The long-term objective is to develop NeuronlQ as an open- source, modular system of multi-functionalities while keeping its interface simple and easy to use for neuroscience researchers.

Landscapes

  • Engineering & Computer Science (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • General Physics & Mathematics (AREA)
  • Multimedia (AREA)
  • Theoretical Computer Science (AREA)
  • Microscoopes, Condenser (AREA)

Abstract

L'invention concerne des méthodes et des appareils permettant l'analyse automatisée d'images neuronales tridimensionnelles acquises par des techniques de microscopie par fluorescence, telles qu'une microscopie de scannage laser confocal et une microscopie de scannage laser multi-photons. Ces méthodes et appareils peuvent être utilisés dans la segmentation et la détection de dendrites et d'épines dendritiques et dans l'extraction de caractéristiques importantes biologiquement, notamment, le nombre, la longueur et le volume de dendrites, le nombre et la densité d'épines, leur volume, forme, type et similaire.
PCT/US2006/019589 2005-05-19 2006-05-19 Methodes et systemes d'analyse d'images neuronales microscopiques tridimensionnelles WO2006125188A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US68251005P 2005-05-19 2005-05-19
US60/682,510 2005-05-19

Publications (1)

Publication Number Publication Date
WO2006125188A1 true WO2006125188A1 (fr) 2006-11-23

Family

ID=36950526

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/019589 WO2006125188A1 (fr) 2005-05-19 2006-05-19 Methodes et systemes d'analyse d'images neuronales microscopiques tridimensionnelles

Country Status (1)

Country Link
WO (1) WO2006125188A1 (fr)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2557525A1 (fr) 2011-08-08 2013-02-13 Instytut Biologii Doswiadczalnej IM.M. Nenckiego Pan Procédé et système pour traiter une image comprenant des épines dendritiques
CN104331892A (zh) * 2014-11-05 2015-02-04 南京理工大学 一种基于形态学的神经元识别与分析方法
ES2560542R1 (es) * 2014-08-19 2016-03-03 Universidad Rey Juan Carlos Método de reconstrucción con un dispositivo háptico de espinas dendríticas
EP3100237A4 (fr) * 2014-01-31 2017-08-23 Afraxis Holdings, Inc. Analyse d'image anatomique distribuée
WO2019022925A1 (fr) * 2017-07-22 2019-01-31 Intelligent Virus Imaging Inc. Procédé d'étude ontologique non supervisée automatisée d'aspects structuraux dans des micrographes électroniques
WO2019032723A1 (fr) * 2017-08-09 2019-02-14 Allen Institute Systèmes, dispositifs et procédés de traitement d'image pour générer une image présentant un marquage prédictif
RU2692038C2 (ru) * 2014-05-19 2019-06-19 Конинклейке Филипс Н.В. Визуализация представляющей интерес ткани в данных контрастированного изображения
DE102020202610A1 (de) 2020-02-28 2021-09-02 Gottfried Wilhelm Leibniz Universität Hannover Verfahren und Vorrichtung zur automatisierten mikroskopischen Analyse von nervenzellhaltigen Proben
US20210407085A1 (en) * 2019-03-29 2021-12-30 Fujifilm Corporation Cell evaluation device, operation method for cell evaluation device, and operation program for cell evaluation device
WO2022076934A1 (fr) * 2020-10-09 2022-04-14 Arizona Board Of Regents On Behalf Of Arizona State University Protection anti-fraude à l'aide de dendrites
US11455333B1 (en) 2021-04-07 2022-09-27 Protochips, Inc. Systems and methods of metadata and image management for reviewing data from transmission electron microscope (TEM) sessions
WO2022216970A1 (fr) * 2021-04-07 2022-10-13 Protochips, Inc. Systèmes et procédés de gestion de métadonnées et d'images pour examiner des données provenant de sessions de microscope électronique à transmission (tem)
US12008764B2 (en) 2023-02-16 2024-06-11 Allen Institute Systems, devices, and methods for image processing to generate an image having predictive tagging

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020004632A1 (en) * 2000-04-10 2002-01-10 Lindquist W. Brent System and method for determining neuronal morphology and effect of substances thereon

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020004632A1 (en) * 2000-04-10 2002-01-10 Lindquist W. Brent System and method for determining neuronal morphology and effect of substances thereon

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE INTERNET W.B. LINDQUIST ET AL:: "3DMA-Neuron: A software package for automated neuronal morphology", XP002398996, retrieved from HTTP://WWW.AMS.SUNYSB.EDU/~LINDQUIS/3DMA/3DMA_NEURON/3DMA_NEURON.HTML *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013021001A1 (fr) 2011-08-08 2013-02-14 Instytut Biologii Doswiadczalnej Im. M. Nenckiego Pan Procédé et système de traitement d'une image comprenant des épines dendritiques
EP2557525A1 (fr) 2011-08-08 2013-02-13 Instytut Biologii Doswiadczalnej IM.M. Nenckiego Pan Procédé et système pour traiter une image comprenant des épines dendritiques
EP3100237A4 (fr) * 2014-01-31 2017-08-23 Afraxis Holdings, Inc. Analyse d'image anatomique distribuée
RU2692038C2 (ru) * 2014-05-19 2019-06-19 Конинклейке Филипс Н.В. Визуализация представляющей интерес ткани в данных контрастированного изображения
ES2560542R1 (es) * 2014-08-19 2016-03-03 Universidad Rey Juan Carlos Método de reconstrucción con un dispositivo háptico de espinas dendríticas
CN104331892B (zh) * 2014-11-05 2017-04-19 南京理工大学 一种基于形态学的神经元识别与分析方法
CN104331892A (zh) * 2014-11-05 2015-02-04 南京理工大学 一种基于形态学的神经元识别与分析方法
WO2019022925A1 (fr) * 2017-07-22 2019-01-31 Intelligent Virus Imaging Inc. Procédé d'étude ontologique non supervisée automatisée d'aspects structuraux dans des micrographes électroniques
WO2019032723A1 (fr) * 2017-08-09 2019-02-14 Allen Institute Systèmes, dispositifs et procédés de traitement d'image pour générer une image présentant un marquage prédictif
AU2018313841B2 (en) * 2017-08-09 2023-10-26 Allen Institute Systems, devices, and methods for image processing to generate an image having predictive tagging
US20210407085A1 (en) * 2019-03-29 2021-12-30 Fujifilm Corporation Cell evaluation device, operation method for cell evaluation device, and operation program for cell evaluation device
DE102020202610A1 (de) 2020-02-28 2021-09-02 Gottfried Wilhelm Leibniz Universität Hannover Verfahren und Vorrichtung zur automatisierten mikroskopischen Analyse von nervenzellhaltigen Proben
DE102020202610B4 (de) 2020-02-28 2021-09-23 Gottfried Wilhelm Leibniz Universität Hannover Verfahren und Vorrichtung zur automatisierten mikroskopischen Analyse von nervenzellhaltigen Proben
WO2022076934A1 (fr) * 2020-10-09 2022-04-14 Arizona Board Of Regents On Behalf Of Arizona State University Protection anti-fraude à l'aide de dendrites
US11455333B1 (en) 2021-04-07 2022-09-27 Protochips, Inc. Systems and methods of metadata and image management for reviewing data from transmission electron microscope (TEM) sessions
WO2022216970A1 (fr) * 2021-04-07 2022-10-13 Protochips, Inc. Systèmes et procédés de gestion de métadonnées et d'images pour examiner des données provenant de sessions de microscope électronique à transmission (tem)
US11755639B2 (en) 2021-04-07 2023-09-12 Protochips, Inc. Systems and methods of metadata and image management for reviewing data from transmission electron microscope (TEM) sessions
US12008764B2 (en) 2023-02-16 2024-06-11 Allen Institute Systems, devices, and methods for image processing to generate an image having predictive tagging

Similar Documents

Publication Publication Date Title
WO2006125188A1 (fr) Methodes et systemes d'analyse d'images neuronales microscopiques tridimensionnelles
US11803968B2 (en) Automated stereology for determining tissue characteristics
Chakraborty et al. Modified cuckoo search algorithm in microscopic image segmentation of hippocampus
Rodriguez et al. Automated three-dimensional detection and shape classification of dendritic spines from fluorescence microscopy images
Kervrann et al. A guided tour of selected image processing and analysis methods for fluorescence and electron microscopy
Rodriguez et al. Three-dimensional neuron tracing by voxel scooping
US11080830B2 (en) Systems and methods for segmentation and analysis of 3D images
EP2948897B1 (fr) Procédé et systèmes pour le comptage de points fish au niveau des cellules
Zhang et al. Dendritic spine detection using curvilinear structure detector and LDA classifier
Mikula Progress towards mammalian whole-brain cellular connectomics
Guan et al. NeuroSeg: automated cell detection and segmentation for in vivo two-photon Ca 2+ imaging data
Mouton et al. Unbiased estimation of cell number using the automatic optical fractionator
Rittscher et al. Microscopic image analysis for life science applications
Hodneland et al. Automated detection of tunneling nanotubes in 3D images
Martinez et al. Impartial: Partial annotations for cell instance segmentation
Srinivasan et al. Automated axon tracking of 3D confocal laser scanning microscopy images using guided probabilistic region merging
US20220058369A1 (en) Automated stereology for determining tissue characteristics
Sanders et al. Learning-guided automatic three dimensional synapse quantification for drosophila neurons
Sui et al. A counting method for density packed cells based on sliding band filter image enhancement
Xu et al. A computer-based system to analyze neuron images
Mabaso Automatic approach for spot detection in microscopy imaging based on image processing and statistical analysis
Štajduhar Computational histology image analysis of human cerebral cortex cytoarchitectonics
Haddar et al. A New Method for Microscopy Image Segmentation Using Multi-scale Line Detection
Suresh et al. Quantification and segmentation of progenitor cells in Time-lapse microscopy
KAKULAPATI et al. A Framework base analyzing cancer cells in blood Image segmentation by using convolution neural networks.

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 06770747

Country of ref document: EP

Kind code of ref document: A1