JUMP TO:


3D tissue imaging and cytometry

Goal:
Understand the kidney in health and disease via 3D imaging and cytometric analyses.

The following image types within this technology are available in the Kidney Tissue Atlas:

  • Composite 3D 8-channel immunofluorescence image volume
    3D volume completely represented as a stack of individual, 8-channel images. Every focal plane image and every channel can be independently inspected.
  • Composite max projection of 8-channel immunofluorescence image volume
    8-channel volume combined into a single maximum projection; composite image consists of 8 channels.
  • RGB max projection of 2-channel (autofluorescence and second harmonic generation) image volume
    Projection of 3D volume collected prior to labeling; channels cannot be controlled.
  • RGB max projection of 8-channel immunofluorescence image volume
    8-channel volume combined into a single maximum projection and converted to RGB color space.

Protocol(s)

Metadata standards


CODEX (CO-detection by InDEXing)

Goal:
To build a library of high-resolution phenotypical maps of kidney biopsies with anchor, immune, and functional markers for in situ spatial analysis at single cell resolution in normal and pathological conditions.

Protocol(s)

Metadata standards

  • Not yet available


Imaging mass cytometry

Goal:
To generate high-dimensional spatially preserved data at single cell resolution to define the cellular milieu and cell-cell interactions in normal and injured kidneys.

Figure 1. Schematic representation of IMC (left) with a reconstruction of the tubular cell make-up of a human cortical kidney section performed using IMC (each color represents a distinct cell type (PT=magenta, TAL=yellow, DCT=cyan).

Protocol(s)

Metadata standards


MULTIMODAL IMAGING MASS SPECTROMETRY

Goal: 
Our multimodal molecular imaging pipeline combines imaging mass spectrometry (IMS), Matrix-assisted laser desorption/ionization (MALDI) IMS, generates molecular images of metabolites and lipids with high spatial fidelity, showing localization to different functional tissue units (FTU) in the kidney. Analytes  can be identified with a combination of accurate mass measurements (<1 ppm error) and complimentary liquid chromatography (LC)-based fragmentation experiments. By integrating this technology with advanced machine learning, Kidney Precision Medicine Project (KPMP) researchers will enable a more comprehensive understanding of the molecular and structural drivers of acute kidney injury (AKI) and chronic kidney disease (CKD).

Figure 1: Multimodal Imaging Mass Spectrometry workflow developed by Vanderbilt University: (A) Autofluorescence, MALDI IMS and stained microscopy data collection is performed on a single tissue section. (B) The data are processed by in-house developed tools. (C) The data are mined and analyzed using our computational workflows.

Protocol(s)

Protocols used by Vanderbilt University TIS for Multimodal Molecular Imaging of Human Kidney Biopsies

Overall Pipeline:

  • VU TIS Multimodal Molecular Imaging Pipeline for KPMP Biopsy Interrogation (not yet available)
  • Tissue Screening and Assessment (not yet available)

Sample Interrogation Assays:

Data Pre-processing and Analysis

Metadata standards

  • Not yet available

Imaging

JUMP TO:


Single-nucleus RNA-seq

Goal:
Identify cell types and states associated with normal and injured kidney functions using gene expression profiling. Identify marker genes for cell type/states and any proportion shifts underlying pathology.

Protocols:

Metadata standards

Single-cell RNA-seq

Goal:
Empirically derive cell subtypes and cell-type-specific gene expression profiles​.

Protocol(s)


Metadata standards


Regional transcriptomics

Goal:
Generate deep transcriptomic signatures from nephron segments defined spatially by antibody staining using laser microdissection.

Protocol(s)

Metadata standards


Spatial transcriptomics

Goal:
Capture whole transcriptome mRNA expression with localization to kidney cells and structures.

Protocol(s)

Metadata standards

  • Not yet available


Single-nucleus (sn) RNA-seq + snATAC-seq (10X Multiome)

Goal:
10X Genomics multiome protocol generates a molecular atlas of the human kidney with comprehensive cell types and minimal processing artifacts. 10X Multiome snATAC-seq + Gene Expression assay allows measurements of chromatin accessibility and transcription from the same cell. This allows direct insights into putative transcription factors or SNPs associated with open chromatin and impact on gene expression.

PROTOCOLS:

METADATA STANDARDS

Transcriptomics

JUMP TO:


Regional proteomics

Goal:
Characterize the kidney proteome in health and disease to identify protein markers that reflect each segment, matrix, and cell type contained in the tissue.

Protocol(s)

Metadata standards

  • Not yet available

Proteomics

JUMP TO:


Spatial metabolomics

Goal:
Localize the small metabolite markers in tissue sections from human kidneys.

Protocol(s)

Metadata standards


Spatial lipidomics

Goal:
Localize the lipid markers in tissue sections from human  kidneys.

Protocol(s)

Metadata standards


Spatial N-glycomics

Goal:
Localize the N-glycan markers in tissue sections from human  kidneys

Protocol(s)

Metadata standards

Metabolomics

JUMP TO:


CUT&RUN

Goal:

To obtain genome-wide profiles of histone post-translational modifications that define chromatin states of healthy and injured/repairing kidney tissue using Cleavage Under Targets and Release Using Nuclease (CUT&RUN). This technology enables the interrogation of chromatin profiles with limited tissue, such as kidney biopsies. 

PROTOCOLS:

METADATA STANDARDS


DNA METHYLATION SEQUENCING

Goal:

The goal of this technology is to uncover methylation signatures of kidney sub-segments that are key determinants of kidney disease progression. This work will result in a comprehensive set of methylation patterns and genotypes to map back to a kidney atlas. This will allow the characterization of methylation signatures of over 20 million CpG sites in individual kidney compartments from both nephrectomies and biopsies using laser microdissection (LMD) methodology with downstream whole-genome bisulfite-sequencing (WGBS).

Workflow: 1) Cryosectioning, 2) LMD processing, 3) DNA isolation, bisulfate conversion, WGBS-sequencing, 4) aligning with the reference genome with Bismark, 5) data analysis in R, and genome program visualization, 6) raw and analyzed data are uploaded to the KPMP data lake.

PROTOCOLS:

METADATA STANDARDS

Epigenetics

JUMP TO:


3D tissue imaging and cytometry

Goal:
Understand the kidney in health and disease via 3D imaging and cytometric analyses.

The following image types within this technology are available in the Kidney Tissue Atlas:

  • Composite 3D 8-channel immunofluorescence image volume
    3D volume completely represented as a stack of individual, 8-channel images. Every focal plane image and every channel can be independently inspected.
  • Composite max projection of 8-channel immunofluorescence image volume
    8-channel volume combined into a single maximum projection; composite image consists of 8 channels.
  • RGB max projection of 2-channel (autofluorescence and second harmonic generation) image volume
    Projection of 3D volume collected prior to labeling; channels cannot be controlled.
  • RGB max projection of 8-channel immunofluorescence image volume
    8-channel volume combined into a single maximum projection and converted to RGB color space.

Protocol(s)

Metadata standards


CODEX (CO-detection by InDEXing)

Goal:
To build a library of high-resolution phenotypical maps of kidney biopsies with anchor, immune, and functional markers for in situ spatial analysis at single cell resolution in normal and pathological conditions.

Protocol(s)

Metadata standards

  • Not yet available


Imaging mass cytometry

Goal:
To generate high-dimensional spatially preserved data at single cell resolution to define the cellular milieu and cell-cell interactions in normal and injured kidneys.

Figure 1. Schematic representation of IMC (left) with a reconstruction of the tubular cell make-up of a human cortical kidney section performed using IMC (each color represents a distinct cell type (PT=magenta, TAL=yellow, DCT=cyan).

Protocol(s)

Metadata standards


MULTIMODAL IMAGING MASS SPECTROMETRY

Goal: 
Our multimodal molecular imaging pipeline combines imaging mass spectrometry (IMS), Matrix-assisted laser desorption/ionization (MALDI) IMS, generates molecular images of metabolites and lipids with high spatial fidelity, showing localization to different functional tissue units (FTU) in the kidney. Analytes  can be identified with a combination of accurate mass measurements (<1 ppm error) and complimentary liquid chromatography (LC)-based fragmentation experiments. By integrating this technology with advanced machine learning, Kidney Precision Medicine Project (KPMP) researchers will enable a more comprehensive understanding of the molecular and structural drivers of acute kidney injury (AKI) and chronic kidney disease (CKD).

Figure 1: Multimodal Imaging Mass Spectrometry workflow developed by Vanderbilt University: (A) Autofluorescence, MALDI IMS and stained microscopy data collection is performed on a single tissue section. (B) The data are processed by in-house developed tools. (C) The data are mined and analyzed using our computational workflows.

Protocol(s)

Protocols used by Vanderbilt University TIS for Multimodal Molecular Imaging of Human Kidney Biopsies

Overall Pipeline:

  • VU TIS Multimodal Molecular Imaging Pipeline for KPMP Biopsy Interrogation (not yet available)
  • Tissue Screening and Assessment (not yet available)

Sample Interrogation Assays:

Data Pre-processing and Analysis

Metadata standards

  • Not yet available

Imaging

Please Upgrade Your Browser.

Unfortunately, Internet Explorer is an outdated browser and we do not currently support it. To have the best browsing experience, please upgrade to Microsoft Edge or Google Chrome.

Upgrade