Fact-checked by Grok 2 weeks ago

Laser capture microdissection

Laser capture microdissection (LCM) is a molecular biology technique that enables the precise isolation of specific cells or cell populations from heterogeneous tissue samples under direct microscopic visualization, using a focused low-energy infrared laser to adhere target cells to a thermoplastic film for downstream analyses such as DNA, RNA, or protein extraction. Developed in 1996 by researchers at the National Cancer Institute, including Michael R. Emmert-Buck, William M. Bonner, and Lance A. Liotta, LCM addressed the longstanding challenge of separating pure cell types from complex tissues without contamination, revolutionizing studies in oncology and other fields by allowing targeted molecular profiling of normal, precancerous, or diseased cells. The principle of LCM involves placing a tissue section on a glass slide coated with a film, visualizing the sample via an , and activating the (typically at 810 nm) to briefly melt the film beneath selected cells, causing them to adhere while surrounding areas remain unaffected; the captured cells can then be catapulted into a collection tube using a focused or collected by gravity in advanced systems. This non-contact method minimizes damage to biomolecules and preserves cellular morphology, distinguishing it from earlier manual microdissection techniques that were labor-intensive and prone to contamination. Complementary variants include (UV) cutting systems, which ablate unwanted tissue around targets using a 355 nm without a transfer film, offering higher for smaller samples like single cells or organelles, though they may introduce more thermal effects. Historically, the concept of laser microdissection originated in the 1970s through pathologists' development of the first systems for excision, but LCM's capture innovation in 1996 marked a pivotal advancement for molecular applications, as detailed in the foundational publication. Subsequent commercial systems, such as those from and , integrated upright or inverted microscopes with pressure catapulting or gravity collection, enhancing efficiency and expanding usability across fixed, frozen, or live s. LCM's applications span , where it isolates neurons for studying neurodegeneration in conditions like ; , enabling proteomic and genomic analyses of tumor microenvironments; forensics for from trace tissues; and plant biology for dissecting specific cell types in complex organs. Its advantages include high specificity (down to single cells), compatibility with various downstream assays like qPCR, microarrays, and , and reduced processing time compared to enzymatic or methods, though it requires specialized costing over $100,000 and trained operators to optimize tissue preparation and avoid artifacts from fixation or . Overall, LCM remains a cornerstone tool in precision medicine and research, facilitating insights into cellular heterogeneity that were previously unattainable.

Definition and History

Definition and Purpose

Laser capture microdissection (LCM) is a that enables the precise isolation of specific populations from heterogeneous sections under direct microscopic , utilizing a to procure targeted cells while adhering to a thermoplastic film for transfer. This method employs either or lasers to either capture or ablate cells, preserving the morphological and molecular integrity of the procured material. The primary purpose of LCM is to facilitate downstream molecular analyses, such as , transcriptomics, , and , on pure subpopulations of cells extracted from complex tissues, including tumors where cellular heterogeneity can confound tissue studies. By isolating specific cells, LCM addresses the limitations of traditional analysis methods, which average signals across diverse cell types and obscure subtype-specific molecular profiles. Key prerequisites for LCM include the use of for visualization and technology for precise selection and capture, distinguishing it from earlier manual microdissection approaches like scraping, which were prone to and required extensive dexterity. Developed to overcome these issues, LCM provides a rapid, one-step process that minimizes extraneous material transfer, ensuring high-purity samples for reliable molecular interrogation.

Historical Development

Laser capture microdissection (LCM) was invented in 1996 by a team at the (NIH), led by Michael R. Emmert-Buck, Robert F. Bonner, Philip D. Smith, Rodrigo Z. Chuaqui, Zhengping Zhuang, Steven R. Goldstein, and Lance A. Liotta, who developed the first (IR) laser-based system to precisely isolate targeted cells from heterogeneous tissue samples under microscopic visualization. This innovation addressed a critical need in for obtaining pure cell populations without contamination from surrounding tissues, marking a significant advancement over manual dissection methods. The technology was patented by the NIH shortly thereafter and exclusively licensed to Engineering (now part of ), which commercialized the inaugural PixCell system in the late 1990s, enabling widespread laboratory adoption. In the early 2000s, LCM evolved with the introduction of (UV) techniques, complementing the original IR capture method and allowing for more versatile sample procurement in systems like the Microlaser Technologies platforms and hybrid IR/UV instruments such as the . These developments expanded LCM's applicability to diverse types and reduced times, fostering into high-throughput workflows. By the 2010s, further refinements included robotic for enhanced and , as seen in advanced systems that automated selection and capture, minimizing user variability. A pivotal milestone came in 2006 with the publication of a standardized protocol in Nature Protocols, which detailed best practices for LCM sample preparation, capture, and downstream molecular analysis, promoting consistency across research labs worldwide. From 2020 to 2025, LCM advanced toward greater automation and compatibility with multi-omics approaches, incorporating features like AI-assisted targeting and seamless integration with and pipelines to support single-cell studies in cancer and . These innovations have driven widespread adoption, with over 7,000 publications indexed in by 2025, underscoring LCM's enduring impact on biomedical research.

Principles of Operation

Basic Mechanism

Laser capture microdissection (LCM) fundamentally operates by using a low-energy to selectively isolate targeted cells from a heterogeneous while preserving their structural and molecular integrity. The core process begins with placing a thin histological on a , overlaid by a transparent film, such as . Under microscopic visualization, an operator identifies and marks cells of interest based on morphological or criteria. A focused pulse is then directed at these cells, activating to adhere specifically to the targeted area without affecting adjacent . Upon removal of , the selected cells are transferred intact to a collection vessel for downstream analysis. The technique employs two primary modes of operation: laser capture and laser cutting. In the laser capture mode, an infrared (IR) laser, typically at a wavelength of 810 nm, delivers photothermal energy to transiently melt the thermoplastic film overlying the selected cells. This localized melting, confined to spot sizes as small as 7.5 μm, causes the polymer to expand and bond firmly to the cell membranes, enabling precise adhesion without direct tissue ablation. In contrast, the laser cutting mode utilizes an ultraviolet (UV) laser, often at 355 nm, to ablate and sever non-target tissue surrounding the cells of interest, followed by a catapulting mechanism—such as a low-pressure air pulse or additional laser pulse—to propel the isolated fragment into a collection cap. These modes can be combined in hybrid systems for enhanced efficiency. A key advantage of LCM is the minimal laser exposure and heat generation, which safeguards biomolecular content. The IR laser's energy is predominantly absorbed by the thermoplastic film, resulting in temperatures that do not exceed levels damaging to RNA, DNA, or proteins in the cells; for instance, the process maintains ambient conditions with transient localized heating below 90°C. Similarly, UV cutting limits collateral damage through precise beam focusing and short pulse durations (e.g., 1 nanosecond), preventing degradation or cross-contamination. This preservation enables reliable extraction of high-quality nucleic acids and proteins from the captured cells. The step-by-step adhesion and lift-off in the capture mode proceeds as follows: (1) the section is apposed to the film; (2) the laser pulse melts the film locally over the target cells; (3) the softened conforms to and engulfs the cellular structures; (4) upon cooling, the film solidifies, securing the cells; and (5) gentle peeling of the film lifts the adhered cells away, leaving surrounding undisturbed. This non-contact ensures high purity and yield, typically capturing hundreds to thousands of cells in minutes.

Laser Physics and Types

Laser energy in laser capture microdissection (LCM) is delivered through a highly focused , enabling precise targeting of cellular structures with minimal impact on surrounding . The typically operates with durations ranging from 0.1 to 10 milliseconds and power levels between 10 and 100 milliwatts, allowing for controlled or activation while maintaining spatial accuracy down to the scale of individual s. This focused delivery relies on the principles of optical focusing, where the spot size determines the resolution, often achieving 1-10 micrometers to match typical dimensions. Infrared (IR) lasers, such as those operating at 810 nanometers (e.g., diode-pumped systems), are commonly employed for non-destructive capture in LCM. These wavelengths are absorbed primarily by a film overlaying the sample, causing localized and to the target cells without direct heating or damage. The E delivered to the capture area is calculated as E = \frac{P \times t}{A}, where P is the , t is the duration, and A is the area, ensuring efficient with . Systems like the XT utilize this IR approach to preserve cellular integrity for downstream molecular analysis. Ultraviolet (UV) lasers, typically at 355 nanometers (e.g., frequency-tripled :YAG), facilitate direct cutting and excision of target regions through photochemical , which breaks molecular bonds without significant thermal effects. This "cold" minimizes to adjacent cells, making UV suitable for in dense tissues. In hybrid systems, UV cutting is often combined with IR capture to optimize both and . LCM lasers are predominantly pulsed to control energy deposition and reduce heat accumulation, though continuous wave (CW) modes may be used in specialized trapping applications for stable cell manipulation. Many modern LCM platforms integrate with confocal microscopy, enabling real-time fluorescence-guided targeting and three-dimensional visualization during dissection. Recent advancements in the include lasers, which employ ultrashort pulses (on the order of 10^{-15} seconds) to further reduce and enable single-cell or even organelle-level from living . These developments enhance and viability, expanding LCM applications in dynamic biological studies.

Procedure

Sample Preparation

Sample preparation for laser capture microdissection (LCM) begins with tissue fixation to preserve cellular structure and molecular content. are commonly fixed using formalin-fixed paraffin-embedded (FFPE) methods for archival stability or fresh frozen techniques to maintain integrity, with sections typically cut to 5-10 μm thickness using a or . Fresh frozen sections are preferred for and protein analyses due to reduced degradation compared to FFPE, though both support high-quality LCM when properly handled. Staining protocols enhance visualization of target cells without compromising downstream analyses. Hematoxylin and eosin (H&E) staining is routinely applied to delineate morphological features, while (IHC) targets specific protein markers for precise cell identification. Over-staining must be minimized to avoid molecular cross-linking or laser interference, with optimized protocols limiting exposure times to under 5 minutes for IHC to preserve integrity. These steps are performed under RNase-free conditions to protect nucleic acids. Sections are mounted on specialized slides compatible with the LCM system's laser type. For ultraviolet (UV) laser systems, polyethylene naphthalate (PEN) membrane slides are used to facilitate precise cutting and capture. Infrared (IR) systems employ slides coated with a transfer film, enabling gentle and release during dissection. Post-staining, dehydration through graded series (e.g., 75%, 95%, 100%) followed by clearing and air-drying is essential to remove water, stabilize , and prevent biomolecular during laser exposure. These steps, completed rapidly within 10-15 minutes, ensure tissue integrity for efficient capture. Live-cell LCM is possible using systems that isolate viable cells without fixation, such as UV laser methods with gravity transfer, suitable for dynamic studies in animal and plant tissues. Quality assessment precedes microdissection, involving microscopic inspection to verify section flatness, to the slide, and uniform distribution, which directly impact capture precision and yield. Non-flat or uneven sections are discarded to avoid artifacts in downstream molecular extractions.

Microdissection Process

The microdissection process in laser capture microdissection (LCM) begins with visualization of the tissue sample under an , typically using brightfield illumination for unstained sections or for stained or labeled specimens to identify specific cell types or regions of interest (ROI). This step allows precise targeting of subpopulations within heterogeneous tissues, such as tumor cells amid surrounding . Target selection follows, where operators manually outline ROIs using a , , or software annotation tools displayed on the live microscopic image, enabling the demarcation of single cells, cell clusters, or larger areas for isolation. Modern systems incorporate semi-automated or AI-assisted selection algorithms to enhance accuracy and speed, particularly for repetitive or complex patterns. Once selected, the laser is activated to fire short pulses that either adhere target cells to a film on a collection cap (in infrared-LCM) or cut around them for (in ultraviolet-LCM), typically requiring multiple pulses to ensure complete procurement without damaging adjacent . The collection cap is positioned directly over the slide during firing, facilitating immediate of the isolated material. A typical LCM session yields hundreds to thousands of cells, depending on the spot size, density, and duration, with higher numbers achievable through repeated captures. Since around 2015, automation software integrated into commercial systems has supported high-throughput workflows, allowing of multiple ROIs to increase efficiency in large-scale studies. Post-capture verification involves the collection cap under low (up to 20x) to confirm the presence of cells, assess purity, and remove any extraneous debris, ensuring the isolated material is suitable for downstream .

Post-Capture Handling

Following the microdissection step, captured cells or fragments are promptly transferred to secure collection vessels to prevent loss and maintain integrity. Common methods include adhering the isolated material to films or caps, which are then placed onto microcentrifuge tubes containing buffers or directly into extraction kits for immediate processing. This transfer is often facilitated by pressure catapulting, ensuring minimal mechanical disturbance. To preserve biomolecular content, especially RNA, which is prone to rapid degradation at room temperature due to RNase activity, samples are either lysed immediately or frozen at -80°C. Lysis inactivates RNases and stabilizes nucleic acids, while cryogenic storage halts enzymatic activity for later analysis. Quality assurance begins with post-capture imaging to verify cell count and morphological integrity, often using the microscope's visualization capabilities. Contamination checks involve assessing surrounding tissue remnants or foreign cells through microscopic inspection and downstream purity assays. Advanced systems integrate for automated, sterile handling, reducing and risks. Microfluidic interfaces have been incorporated in some systems to minimize sample during , enhancing yield for low-input analyses. These steps culminate in the initiation of , setting the stage for molecular extraction while safeguarding sample viability.

Molecular Extraction

Nucleic Acid Extraction

Following laser capture microdissection (LCM), nucleic acid extraction begins with cell lysis, typically using specialized kits optimized for low-input samples to preserve integrity and yield. For DNA isolation, the Arcturus PicoPure DNA Extraction Kit is commonly employed, providing a streamlined proteinase K-based procedure that yields PCR-ready DNA from as few as 10 captured cells without requiring spin columns. This method typically recovers 50-300 ng of total DNA from LCM samples, sufficient for downstream applications like PCR amplification, though actual yields depend on cell type and tissue source. RNA extraction from LCM-captured cells prioritizes integrity to support sensitive analyses such as reverse transcription PCR (RT-PCR) and RNA sequencing (RNA-seq). The RNeasy Micro Kit from QIAGEN is widely used, employing silica-membrane column-based purification to isolate high-quality total RNA from as little as one cell, with yields up to 45 µg possible from microdissected material. A critical step involves on-column DNase I treatment to eliminate genomic DNA contamination, ensuring RNA purity (A260/A280 ratio >1.8) essential for accurate transcriptomic profiling. To optimize yields, extraction buffer volumes are scaled proportionally to the number of captured cells, often using 10-50 µL of for 100-1,000 cells to minimize dilution and maximize recovery efficiency. In formalin-fixed paraffin-embedded (FFPE) samples processed via LCM, DNA yields fragmented products averaging 200-500 bp in size due to fixation-induced crosslinks and degradation, necessitating short-amplicon or targeted sequencing strategies. Recent advancements include single-cell LCM coupled with protocols, such as immuno-LCM-RNAseq, which integrate immunofluorescence-guided capture with next-generation sequencing (NGS) for spatially resolved transcriptomics from 2022 onward, enabling high-sensitivity analysis of rare cell populations. Post-extraction, quantity and quality are assessed using spectrophotometric methods like NanoDrop for rapid UV absorbance-based measurement (A260) or fluorometric assays like for higher specificity and sensitivity in low-concentration samples (<10 ng/µL), confirming suitability for downstream NGS library preparation.

Protein and Metabolite Extraction

Protein extraction from laser capture microdissection (LCM) samples typically involves using buffers containing detergents such as 2% in 300 mM Tris-HCl (pH 8.0) to solubilize proteins effectively while preserving integrity for downstream applications. This approach includes boiling at 99°C for 25 minutes followed by and additional heating at 80°C for 2 hours to enhance extraction efficiency, often yielding sufficient material for analysis from limited cell numbers. Extracted proteins are compatible with blotting for targeted validation and (MS) workflows, where reduction with DTT and with prepare samples for enzymatic . Typical yields range from 0.1 to 10 μg of protein from approximately 1,000 cells, depending on type and fixation , enabling proteomic profiling of over 5,000 protein groups. These low yields pose challenges, necessitating highly sensitive detection methods like nano-LC-MS/MS to identify and quantify proteins reliably from microdissected populations. Recent protocols emphasize preservation during LCM, such as optimized fixation and rapid processing to maintain states for in spatial . Integration with platforms, including iTRAQ labeling, allows multiplexed relative quantification of proteins from LCM-isolated cells, as demonstrated in studies of lung adenocarcinoma mitochondrial proteomes. Protein concentrations are commonly quantified using the assay, which measures total protein via colorimetric detection of Cu⁺-BCA complexes formed under alkaline conditions. Metabolite extraction from LCM samples employs organic solvents like /chloroform mixtures, following a modified Bligh-Dyer biphasic protocol to separate polar and non-polar metabolites efficiently. This method involves adding cold solvent to microdissected material, vortexing, and to isolate metabolites for LC-MS , ensuring comprehensive coverage of small molecules. Cold processing throughout extraction, typically at -80°C or on ice, halts enzymatic activity and prevents metabolic turnover, preserving snapshot profiles of cellular states. Similar to protein challenges, low sample amounts demand sensitive platforms; quantification often uses NMR spectroscopy for absolute metabolite levels in microdissected tissues, leveraging signal integration for non-destructive .

Applications

Research Applications

Laser capture microdissection (LCM) plays a pivotal role in research by enabling the isolation of specific populations from heterogeneous tissues, facilitating the of tumor heterogeneity and the of driver mutations in cancer subclones. In studies, LCM combined with low-input whole-genome sequencing has validated spatial subclone structures, confirming PTEN inactivating mutations in (DCIS) and invasive compartments, revealing of distinct subclones. Similarly, in , LCM enhances genomic analysis of and mutations by isolating pure tumor cells from cytology samples, overcoming dilution from non-neoplastic elements and improving next-generation sequencing accuracy. These applications underscore LCM's utility in decoding spatial genomic alterations that drive tumor progression. In , LCM supports spatial mapping of proteins in , particularly for research, by isolating specific neuronal populations for targeted analysis. For instance, LCM has been used to microdissect pyramidal cells from the CA1 region of the in Alzheimer's brains, identifying upregulated proteins such as creatine kinase B-type (CKB), 14-3-3-γ, and heat shock cognate 71 (Hsc71), which may reflect protective or pathological responses in affected neurons. This approach provides insights into region-specific protein alterations, advancing understanding of neurodegenerative mechanisms. Recent studies have leveraged LCM for immune cell profiling in tumors through integration with single-cell RNA sequencing (scRNA-seq). A 2023 analysis employed LCM-seq to spatially profile the , enabling transcriptomic dissection of immune infiltrates and revealing heterogeneity in immune responses within solid tumors. Emerging integrations post-2020 combine LCM with platforms like Visium, as demonstrated in research where LCM-guided whole-genome sequencing complemented Visium data to map spatiotemporal cancer cell trajectories and clonal domains. LCM has had a broad impact on and biomedical investigations. In plant biology, LCM enables the of specific types from complex organs, such as vascular tissues or reproductive structures, for transcriptomic and proteomic analyses to study development and stress responses.

Clinical Applications

Laser capture microdissection (LCM) plays a pivotal role in clinical diagnostics, particularly in precision , where it enables the of specific tumor s from heterogeneous tissue samples for targeted analysis. In formalin-fixed paraffin-embedded (FFPE) biopsies, LCM facilitates the enrichment of neoplastic cells, improving the accuracy of genomic and proteomic profiling to identify actionable mutations and guide personalized treatment decisions. For instance, in diagnostics, LCM isolates tumor cells from surrounding , enhancing the sensitivity of molecular assays for detecting driver mutations such as or ALK alterations. In and forensics, LCM supports cell-specific molecular analysis in complex samples, including autopsies and . Pathologists use LCM to procure pure populations of cells from archived tissues, enabling downstream or sequencing to resolve ambiguous diagnoses or confirm causes of death without contamination from adjacent non-target cells. In forensic applications, the technique isolates individual contributor cells from mixed biological traces, such as on surfaces, allowing for STR profiling and linkage to specific individuals in criminal investigations. Therapeutically, LCM aids in profiling the to inform strategies by dissecting interactions between cancer cells and immune infiltrates. By capturing distinct compartments—such as tumor nests versus stromal regions—LCM reveals expression patterns of immune checkpoints like in non-small cell lung carcinoma. This spatial resolution supports the development of tailored immunotherapies by identifying immunosuppressive elements within the microenvironment. A notable involves LCM's application in detecting (MRD) in hematologic malignancies, such as (CLL). In pseudofollicles, LCM isolates proliferating B-cell clones from surrounding tissues, enabling clonal sequencing to monitor residual leukemic cells post-treatment and assess relapse risk. This approach enhances MRD detection sensitivity, guiding decisions on consolidation therapies like targeted inhibitors. Recent advancements integrate LCM with liquid biopsy enhancements, where microdissected tissue-derived profiles complement analysis for more comprehensive monitoring in 2024 workflows.

Advantages and Limitations

Key Advantages

Laser capture microdissection (LCM) offers exceptional precision in isolating target cells or subcellular components from heterogeneous tissues, achieving resolutions down to 7.5 μm with minimal from adjacent structures. This subcellular targeting surpasses traditional manual methods, which often introduce unwanted cellular material and compromise sample purity, enabling the procurement of homogeneous populations for downstream analyses. The technique's versatility allows its application across diverse sample types, including fixed, frozen, and live tissues, while preserving the spatial context of cellular microenvironments. This adaptability supports a wide range of molecular investigations, from to , without requiring extensive sample preprocessing that could alter native architecture. LCM facilitates rapid processing, capable of isolating thousands of cells within several minutes, which significantly reduces overall analysis time compared to fluorescence-activated cell sorting (FACS) in complex, heterogeneous samples. Additionally, it maintains high molecular fidelity, yielding RNA with integrity numbers (RIN) greater than 7 post-capture, ensuring reliable data for sensitive assays like . Recent advancements as of 2025, including automated systems with enhanced laser precision and integration for multi-omics workflows—such as AI-guided targeting introduced in 2024—have further boosted throughput, allowing efficient extraction of DNA, RNA, and proteins from the same isolated cells while minimizing degradation.

Limitations and Challenges

One major limitation of laser capture microdissection (LCM) is its low yield of biological material, typically limited to small numbers of cells ranging from 10 to per capture session, which often requires subsequent techniques to generate sufficient input for downstream analyses such as next-generation sequencing (NGS). This constraint arises from the precision-focused of the technique, which prioritizes targeted isolation over bulk collection, potentially introducing bias if artifacts occur. The high cost of LCM systems represents another significant challenge, with equipment prices exceeding $100,000 and requiring extensive operator training to ensure accurate use and minimize errors. Additionally, in formalin-fixed paraffin-embedded (FFPE) tissues, RNA degradation remains a persistent issue due to cross-linking during fixation, though improvements in extraction protocols have enhanced recovery and integrity for transcriptomic studies. Tissue curling during sectioning preparation can further complicate captures by distorting sections and reducing adhesion efficiency. Technical challenges include laser-induced artifacts in pigmented samples, where UV-based systems may cause unintended damage from pigment absorption, affecting molecular integrity. AI-assisted targeting helps mitigate these by automating selection and reducing manual errors through overlays. When higher throughput or non-spatial resolution is needed, alternatives like fluorescence-activated (FACS) or emerging spatial technologies may be preferable, as they avoid physical while providing comparable cellular specificity.

Commercial Systems

Leica LMD Systems

Leica Microsystems' laser microdissection (LMD) systems utilize a UV laser to enable precise, contact-free isolation of specific cells or tissue regions from heterogeneous samples, facilitating downstream molecular analyses such as DNA, RNA, or protein extraction. These systems employ gravity-based collection, where dissected material slides into a collection vessel below the sample, minimizing contamination and preserving sample integrity. Key models include the LMD6500 and LMD7000, introduced in 2009 as advanced iterations building on earlier systems like the LMD6000 from 2005. The LMD6500 is optimized for upright configurations, offering reliable for routine applications, while the LMD7000 integrates with inverted for enhanced flexibility in live-cell work. Both models use a nitrogen-purged UV (355 nm) for clean cutting along user-defined contours, with the beam guided via for high precision without stage movement. These systems feature seamless integration with Leica's DM series microscopes, supporting brightfield, , and confocal imaging modes. Accompanying software provides tools for region-of-interest (ROI) selection, automated cutting patterns, and when paired with THUNDER imagers, enabling volumetric analysis of dissected areas. High-throughput capabilities allow for rapid processing, with optimized workflows supporting up to hundreds of dissections per session in multi-well formats. In 2023, updates incorporated AI-driven morphology recognition via the Aivia software's Pixel Classifier, automating ROI identification based on cellular features for more efficient high-throughput applications; as of May 2025, Aivia 15 further enhances these AI tools for integration with LMD systems. These enhancements build on the original LMD technology introduced in 2005, which revolutionized by combining laser precision with user-friendly interfaces. Leica LMD systems excel in high-precision , where they isolate pure cell populations from complex tissues like sections, and support live-cell imaging through climate-controlled stages that maintain viability during . They are particularly suited for applications requiring subcellular , such as discovery in frozen or paraffin-embedded samples. In academia, Leica LMD systems hold a dominant position, especially in research, where they have enabled pioneering studies on single-neuron isolation and mapping by providing contamination-free, targeted extractions from intricate tissues.

MMI Systems

Molecular Machines & Industries (MMI), founded in 1992 in , , specializes in microscope-based systems for micromanipulation and single-cell isolation, including laser microdissection technologies. The company's laser capture microdissection platforms emphasize contamination-free collection and preservation of biomolecular integrity for downstream analyses. In November 2024, MMI entered a strategic partnership with to enhance single-cell solutions, combining MMI's isolation expertise with ZEISS's microscopy capabilities. The primary MMI systems for laser capture microdissection are the CellCut and CellCut Plus, which employ a low-pulse-energy UV laser (355 nm) for precise cutting with sub-micrometer resolution. Unlike ablative methods, these systems use a non-contact, gentle ablation approach followed by collection via adhesive caps (CapSure or CapLift technology), enabling efficient transfer without physical manipulation of the sample. Designed for integration with fixed-stage inverted microscopes, the platforms support automated dissection workflows through software-controlled stage movement and laser pulsing, facilitating high-throughput processing. They accommodate a range of sample types, including formalin-fixed paraffin-embedded (FFPE) sections, fresh frozen cryosections, live cells, cytospins, and thick tissues up to several hundred micrometers via Z-drill functionality. MMI systems are particularly noted for their ability to maintain high RNA integrity during isolation, with extracted RNA from laser-captured samples achieving (RIN) values of 9–10 when using optimized extraction kits like RNeasy Micro. This preservation outperforms some infrared-based alternatives, minimizing degradation for sensitive transcriptomic studies. In applications, the systems enable targeted isolation of diseased cell populations from heterogeneous tissues, supporting integration with quantitative (qPCR) workflows for validation and biomarker discovery. Recent innovations include add-on modules for microfluidic compatibility, such as the CellManipulator, which enhances single-cell handling in environments for downstream analysis. These features extend the platform's utility in spatial and high-resolution single-cell , allowing seamless transfer of isolated material into nanoliter-volume reactors.

System Comparisons

Major commercial laser capture microdissection (LCM) systems, including those from , & Industries (MMI, in strategic partnership with ), and , differ primarily in technology, collection mechanisms, and workflow automation, influencing their suitability for various applications. LMD systems employ a UV coupled to an for precise cutting, with samples collected via gravity drop into a or tube, enabling efficient handling of larger tissue areas without physical contact. In contrast, MMI and PALM MicroBeam systems also utilize UV but incorporate catapulting or contact-free propulsion for sample retrieval, which is particularly advantageous for fragile or live samples to minimize damage. ’s systems combine IR and UV for both capture and cutting, offering versatility for dense tissues but requiring proprietary . Resolution and precision vary slightly across systems, with MMI CellCut achieving repositioning accuracy below 1 μm and step resolution of 0.156 μm, supporting subcellular isolation. Leica LMD and Zeiss PALM systems provide comparable sub-micrometer precision using pulsed UV lasers at 355 nm, suitable for resolutions down to approximately 1 μm in tissue sections. Throughput is higher in Leica LMD systems due to configurable ultra-scanning stages that allow rapid processing of multiple samples, making them ideal for high-volume workflows. MMI systems excel in yield for delicate samples, with cleaner laser cuts reducing contamination and preserving RNA/DNA integrity, though they may process fewer samples per session compared to Leica. All major systems support downstream omics analyses, including and , but LMD demonstrates an edge in proteomics yield due to its efficient capture from heterogeneous tissues. holds a leading position in the 2025 market alongside (in partnership with MMI) and Thermo Fisher, with the overall LCM market valued at approximately USD 203 million as of 2024 and projected to grow at 8.5% CAGR through 2030. Legacy systems like Thermo Fisher’s PixCell, an early IR-based LCM platform, have been largely superseded by newer models but remain referenced for historical workflows in fixed tissues. Emerging alternatives from manufacturers are gaining traction in the region, contributing to market expansion through cost-effective options, though they currently represent a smaller share focused on applications. When selecting an LCM system, consider sample type and budget: LMD suits high-throughput needs for robust tissues like FFPE sections, while MMI/ is preferable for fragile or live samples requiring gentle handling; Thermo Fisher offers balanced automation for integrated pipelines, with overall costs typically ranging from USD 100,000 to 200,000 depending on configuration.

References

  1. [1]
    Dissecting good from bad with laser-capture
    Aug 11, 2023 · Bonner, Ph. D., and Lance Liotta, M.D., Ph. D., invented laser-capture microdissection (LCM) to rapidly and precisely select specific cells ...
  2. [2]
    Laser capture microdissection: from its principle to applications ... - NIH
    LCM, also known as laser microdissection and pressure catapulting, is one of the most powerful and useful techniques in various research areas.
  3. [3]
    An Introduction to Laser Microdissection - Leica Microsystems
    Oct 9, 2024 · Laser microdissection (LMD) is a highly selective process for preparing samples of DNA, RNA, protein, or other biomaterials for analysis.
  4. [4]
    Laser Microdissection - an overview | ScienceDirect Topics
    Laser microdissection (LM) is defined as a technique for selecting and isolating specific cells from fixed or frozen tissues based solely on their ...
  5. [5]
    20 Years of Leica Laser Microdissection | Learn & Share
    May 10, 2021 · The very first Laser Microdissection system was invented already 45 years ago back in 1976 by German pathologists [1,2]. They used an upright ...
  6. [6]
    Laser-capture microdissection | Nature Protocols
    Jun 27, 2006 · Laser-capture microdissection (LCM) is a method to procure subpopulations of tissue cells under direct microscopic visualization.
  7. [7]
  8. [8]
    Laser capture microdissection: Big data from small samples - PMC
    Dec 1, 2015 · In 1996, Emmert-Buck and coworkers at the National Institutes of Health introduced the infrared (IR) laser capture microdissection system [38].
  9. [9]
    Laser capture microdissection - PubMed
    Laser capture microdissection (LCM) is a technique for isolating pure cell populations from a heterogeneous tissue section or cytological preparation via ...
  10. [10]
    Laser Microdissection System - an overview | ScienceDirect Topics
    In 1996, Emmert-Buck and co-workers developed an IR-LCM system to separate target cells from the tissue by pulsing with an infrared laser of approximate 810 nm ...
  11. [11]
    Computer-Aided Laser Dissection: A Microdissection Workflow ... - NIH
    Dec 11, 2018 · To date, there are over 4000 peer-reviewed publications in the literature using laser dissection covering a range of topics, including the ...
  12. [12]
    Advances in Spatial Multi‐Omics: A Review of Multi‐Modal Mass ...
    May 12, 2025 · This review focuses on mass spectrometry-based spatial omics, specifically matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI).
  13. [13]
    laser microdissection - Search Results - PubMed
    - Total number of publications for laser microdissection: **7,618**
  14. [14]
    Laser capture microscopy - PMC - NIH
    This review concerns a recent advance in microdissection techniques, namely laser capture microdissection (LCM).
  15. [15]
    Laser Capture Microdissection in Cutaneous Research
    LCM is a technique that isolates cells of interest or even a single cell from a heterogeneous tissue specimen using a laser source under microscopic ...
  16. [16]
    Laser Capture Microdissection - an overview | ScienceDirect Topics
    The laser energy may be infrared (810 nm) or ultraviolet (355 nm) or used in combination [120]. The isolated cells may then be subjected to DNA, RNA or protein ...Missing: wavelength | Show results with:wavelength
  17. [17]
    Laser Capture Microdissection (LCM) - Thermo Fisher Scientific
    Laser capture microdissection is an automated technique that enables isolation of specific cells from a mixed population under microscopic visualization.
  18. [18]
    Laser capture microdissection for protein and NanoString RNA ...
    Laser energy in the capture method is infrared (810 nm), while in the cutting mode the laser is ultraviolet (355 nm). Infrared lasers melt a thermolabile ...
  19. [19]
    Laser Capture Microdissection of Paraffin-Embedded Tissues
    Laser microdissection was con- ducted on a PixCell II microdissection apparatus, with aperture setting at 30 µm, laser pulse power at 50. mW, and laser pulse ...
  20. [20]
    Laser Capture Microdissection - A Demonstration of the Isolation of ...
    Feb 6, 2015 · The current anatomical resolution is around 5-10 µm as the smallest spot size that can be consistently produced on the LCM caps to isolate a ...
  21. [21]
    Arcturus(XT) infrared capture and UV cutting methods - PubMed - NIH
    Laser capture microdissection (LCM) is a technique that allows the precise procurement of enriched cell populations from a heterogeneous tissue under direct ...Missing: patent | Show results with:patent
  22. [22]
    Laser Capture Microdissection for the Investigative Pathologist
    Nov 13, 2013 · Essentially, there are 2 main types of laser systems: near-IR capture laser and UV-cutting laser. The near-IR capture laser is the original LCM ...
  23. [23]
    [PDF] Arcturus Cellect LCM System - Thermo Fisher Scientific
    The solid-state. UV laser permits superior speed and precision, and is well suited for microdissecting dense tissue structures and rapidly capturing large.
  24. [24]
    Laser capture microdissection: Should an ultraviolet or infrared laser ...
    Aug 15, 2013 · The aim of this study was to objectively compare the two main LCM systems, one based on an ultraviolet (UV) laser and the other based on an infrared (IR) laser.
  25. [25]
    Comparison of Laser Capture Microdissection and Laser ...
    The IR laser helps to capture the cells of interest, and the UV laser micro-dissects the captured cells. Laser Capture Microdissection (LCM), Laser ...Missing: wavelength | Show results with:wavelength
  26. [26]
    Gentle Laser Microdissection for Cell Clusters, Fixed and ... - ZEISS
    Laser microdissection needs to be free of contamination and guarantee preservation of the material. ... Moreover, laser capture microdissection is also a valuable ...Missing: limitations scraping
  27. [27]
    Femtosecond laser microdissection for isolation of regenerating C ...
    We present femtosecond laser microdissection (fs-LM), a single-cell isolation method that dissects specific cells directly from living tissue.
  28. [28]
    (PDF) Laser Capture Microdissection of FFPE Tissue Sections ...
    Aug 6, 2025 · paraffin embedded. Thickness of microtome sections routinely varies between 5 and. 15 mm. With PALM MicroBeam ...
  29. [29]
    [PDF] Sample Preparation For Laser Microdissection
    Important: Read the Instruction Manual for your laser microdissection system and for all other products used in your work before following a protocol.
  30. [30]
    Immunoguided Laser Assisted Microdissection Techniques for DNA ...
    However, when morphological cell characteristics obtained by routine H&E staining do not allow adequate recognition of the target cells, IHC is required and ...Missing: over- | Show results with:over-
  31. [31]
    Optimized protocol to preserve RNA integrity for laser capture ...
    Laser capture microdissection allows researchers to target specific cells, including immunolabeled MEC, and isolate them from adjacent unlabeled cells for their ...
  32. [32]
    Fixation and Laser Capture Microdissection of Plant Tissue for RNA ...
    Jul 24, 2023 · We developed protocols for fixation, embedding, and sectioning of plant tissue followed by laser capture microdissection (LCM) and processing for RNA recovery.
  33. [33]
    Optimizing Frozen Sample Preparation for Laser Microdissection
    Sample preparation is crucial for quality RNA, DNA and protein retrieval, where it often determines the feasibility of a laser microdissection project. The ...
  34. [34]
    Industrialized, Artificial Intelligence-guided Laser Microdissection for ...
    Jun 3, 2022 · This protocol describes a high-throughput workflow, enabled by artificial intelligence (AI), that segments images of hematoxylin and eosin (H&E)- ...<|control11|><|separator|>
  35. [35]
    Laser Capture Microdissection for Protein and NanoString RNA ...
    Laser capture microdissection (LCM) allows the precise procurement of enriched cell populations from a heterogeneous tissue, or live cell culture, under direct ...
  36. [36]
    High-Throughput Microdissection for Next-Generation Sequencing
    In this study, we demonstrate the accuracy and precision of xMD by rapidly isolating immunostained targets, including cytokeratin AE1/AE3, p53, and estrogen ...
  37. [37]
    A Laser Capture Microdissection Protocol That Yields High Quality ...
    Jun 6, 2019 · First, dispense 50 microliters of the lysis buffer containing beta mercaptoethanol into the cap of the collection tube. Lyse the sample by ...
  38. [38]
    Laser Capture Microdissection in the Tissue Biorepository - PMC
    Today, there are two general classes of laser microdissection: infrared (IR) laser-capture system and ultraviolet (UV) laser-cutting system. The laser-capture ...
  39. [39]
    Optimized Method for Robust Transcriptome Profiling of ... - Frontiers
    Jun 12, 2017 · We find that high quality RNA (RNA integrity number, RIN, >9) of sufficient concentration can be isolated from laser-captured material from ...
  40. [40]
    Laser Microdissection | Precise Cell Isolation - Molecular Machines
    The low pulse energy laser (less than 500 ps) creates a very precise cutting edge (up to 0.3 µm) and therefore protects the surrounding area and biological ...
  41. [41]
    Automated Laser Microdissection (LMD) for Spatial Omics Research
    Automated LMD is an advanced technology that enables the precise and fully automated dissection of tissue, single cells, and even subcellular compartments. With ...Missing: 2015 | Show results with:2015
  42. [42]
    Laser capture microdissection and native mass spectrometry for ...
    Mar 7, 2024 · Here, we demonstrate that these challenges may be overcome through integration of laser capture microdissection in the workflow. We show ...
  43. [43]
    [PDF] Laser Capture Microdissection Cells-to-CT
    Oct 2, 2014 · Prepare the lysis buffer and tubes: a. Label a 0.5-mL GeneAmp® Thin-Walled Reaction Tube for each microdissected sample that you are ...
  44. [44]
    Arcturus® PicoPure® DNA Extraction Kit - Thermo Fisher Scientific
    The Arcturus® PicoPure® DNA Extraction Kit offers an easy, streamlined genomic extraction procedure that produces PCR-ready DNA from as few as 10 cells.
  45. [45]
    Laser Capture Microdissection for the Investigative Pathologist
    A typical. LCM sample can provide between 50 and 300 ng total of DNA; however, the DNA will be fragmented due to the formalin fixa- tion. For polymerase chain ...
  46. [46]
    RNeasy Kits | RNA Extraction Kits - QIAGEN
    In stock $40 deliveryRNeasy Kits are reliable RNA extraction kits that can purify high-quality total RNA from cells, tissues, and yeast.
  47. [47]
    [PDF] RNeasy® Micro Kit - QIAGEN
    Add the. DNase I incubation mix (80 µl) directly to the RNeasy MinElute spin column membrane. Place on the benchtop (20–30°C) for 15 min. Add 350 µl Buffer RW1 ...Missing: laser capture treatment
  48. [48]
    A Systematic Comparison of Protocols for Recovery of High-Quality ...
    Oct 1, 2021 · LCM was performed using the PixCell II Laser Capture Microdissection System (Arcturus Engineering, Mountain View, CA, USA). The power setting ...
  49. [49]
    Robust Acquisition of Spatial Transcriptional Programs in Tissues ...
    Here we present a robust method based on immunofluorescence-guided laser capture microdissection (immuno-LCM-RNAseq) to acquire finely resolved transcriptional ...<|separator|>
  50. [50]
    Comparison of DeNovix, NanoDrop and Qubit for DNA quantification ...
    Jun 17, 2024 · We compared three DNA quantification platforms, including two UV spectrophotometry-based techniques (DeNovix and NanoDrop) and one fluorometry-based approach ( ...
  51. [51]
  52. [52]
    Development of a laser capture microscope-based single-cell-type ...
    Jun 1, 2016 · Laser capture microdissection (LCM) is a technique by which individual cells can be harvested from tissue sections by tacking selected cells to ...Tissue Preparation For Lcm... · Protein Extraction From... · Nano Lc-Ms/ms
  53. [53]
    Utilization of Laser Capture Microdissection coupled to Mass ... - NIH
    The method described herein, combining LCM and MS, has been designed to identify the proteome of different cellular protrusions.Missing: phosphoprotein | Show results with:phosphoprotein
  54. [54]
    Laser capture microdissection coupled mass spectrometry (LCM-MS ...
    Jun 17, 2020 · Here we describe a microproteomics technique to analyse H&E-stained, formalin-fixed paraffin-embedded (FFPE) tissues.
  55. [55]
    Laser Capture Proteomics: spatial tissue molecular profiling ... - NIH
    A strength of LCM is tissue spatial molecular profiling of PTMs (phosphoproteins): mapping activated, or down-regulated, kinase pathways. RNA transcripts ...
  56. [56]
    Quantitative proteomics analysis of mitochondrial proteins in lung ...
    Laser capture microdissection (LCM) was used to isolate the target cells ... (iTRAQ) combined with two-dimensional liquid chromatography-tandem mass ...
  57. [57]
    Simultaneous extraction of proteins and metabolites from cells ... - NIH
    Therefore, we use chloroform [4] with the methanol/water mixture to separate the polar and non-polar metabolites efficiently.Missing: LCM | Show results with:LCM
  58. [58]
    Liquid Chromatography-Mass Spectrometry Metabolic and ... - NIH
    Jun 5, 2015 · The dried cell pellet was stored at -80°C until extraction. Samples were kept on ice from collection to metabolite extraction to prevent ...
  59. [59]
    A novel procedure for the quantitative analysis of metabolites ...
    Jun 30, 2011 · We provide a guideline and detailed procedures towards the quantitative analysis of laser micro-dissected (LM) tissues in oilseed rape (Brassica napus).
  60. [60]
    Spatial genomics maps the structure, nature and evolution of cancer ...
    Nov 9, 2022 · In situ mutation detection and visualization of intratumor heterogeneity for cancer research and diagnostics. Oncotarget 4, 2407–2418 (2013) ...
  61. [61]
    Laser capture microdissection in lung cancer: a narrative review
    Key Content and Findings: This narrative review provides an overview of recent years LCM technological innovations regarding the attempt to make it more usable ...<|control11|><|separator|>
  62. [62]
    Laser capture microdissection for biomedical research: towards high ...
    Aug 5, 2023 · This review focuses on the differences among four types of commonly used LCM technologies and their applications in omics and disease research.
  63. [63]
    Microdissected Pyramidal Cell Proteomics of Alzheimer Brain ...
    Nov 18, 2021 · Based on previous findings from experimental studies, CKB and Hsc71 likely exhibit protective effects, whereas 14-3-3-γ may represent a detrimental pathway.
  64. [64]
    Merger of Laser Capture Microdissection and Mass Spectrometry
    This chapter will highlight the use of LCM in the large‐scale proteomic analysis of AD amyloid plaques. Particular focus will be directed to detailing the ...
  65. [65]
    [PDF] A spatiotemporal cancer cell trajectory underlies glioblastoma ...
    May 14, 2025 · Visium spatial transcriptomics (ST) (10X Genomics) (Fig. 1A) ... C) Spatial clonal domains compared across LCM-WGS pyclone deconvolution and Visium ...
  66. [66]
    Laser Capture Microdissection Market Size and Forecast 2025 to 2034
    Aug 4, 2025 · The global laser capture microdissection market size accounted for USD 220.60 million in 2025 and is forecasted to hit around USD 463.14 million ...
  67. [67]
    Whole-slide laser microdissection for tumour enrichment - PubMed
    Abstract. The practical application of genome-scale technologies to precision oncology research requires flexible tissue processing strategies that can be used ...
  68. [68]
    Laser capture microdissection in forensic research: a review - PMC
    Aug 3, 2010 · Laser capture microdissection (LCM) offers a valuable tool for precise separation of specific cells. This review summarises all possible forensic applications ...
  69. [69]
    Deciphering functional tumor-immune crosstalk through highly ...
    Mar 6, 2025 · We introduce mipDVP, an advanced approach integrating highly multiplexed imaging, single-cell laser microdissection, and sensitive mass spectrometry.
  70. [70]
    Laser microdissection for the assessment of the clonal relationship ...
    Aug 6, 2025 · Laser microdissection for the assessment of the clonal relationship between chronic lymphocytic leukemia/small lymphocytic lymphoma and ...
  71. [71]
    Enabling Precision Oncology Through Precision Diagnostics
    Jan 24, 2020 · Detection of minimal residual disease in B lymphoblastic leukemia by high-throughput sequencing of IGH. ... Laser capture microdissection ...
  72. [72]
    Advancing drug development by leveraging microdissection for ...
    May 29, 2024 · Background: Combining laser capture microdissection (LCM) with proteogenomics in cancer research offers a targeted way to way to explore the ...
  73. [73]
    Laser Capture Microdissection in the Spatial Analysis of Epigenetic ...
    Feb 9, 2022 · Laser capture microdissection (LCM) is a powerful technique used to isolate a targeted cell group with extreme microscopic precision.
  74. [74]
    Laser Capture Micro-dissection (LCM) 2025-2033 Analysis
    Rating 4.8 (1,980) Jun 28, 2025 · Technological advancements: Improved laser precision, automated systems, and integration with downstream analytical platforms enhance ...
  75. [75]
  76. [76]
    Laser capture microscopy coupled with Smart-seq2 for precise ...
    Jul 8, 2016 · Here we report a robust and highly efficient strategy for LCM coupled with full-length mRNA-sequencing (LCM-seq) developed for single-cell transcriptomics.
  77. [77]
    Thermo Scientific™ Arcturus Cellect Laser Capture Microdissection ...
    The Cellect supports all consumables and reagents used by Arcturus legacy instruments. $262100.00 - $262100.00. Specifications. For Use With (Equipment), The ...
  78. [78]
    Laser capture microdissection analysis of gene expression ... - PNAS
    We tested the ability of laser capture microdissection (LCM) and real-time quantitative reverse transcription PCR to selectively analyze RNA from lesional ...
  79. [79]
    An Optimized Protocol of Laser Capture Microdissection for Tissue ...
    Sep 17, 2020 · We present a protocol optimized for radish root tissue sections using Steedman's wax embedding to obtain high-quality RNA suitable for next-generation ...
  80. [80]
    Laser Capture Microdissection Market Size & Share Analysis
    Jun 17, 2025 · Regulators now emphasize companion diagnostics, nudging firms to use laser capture microdissection so biomarker assays start with pure cell ...
  81. [81]
    A Comparison of Tissue Dissection Techniques for Diagnostic ...
    Jun 6, 2023 · Unlike IR-LCM, the entire tissue field is irradiated, allowing the EVA film to selectively melt directly over the pigmented cells. The pigmented ...<|control11|><|separator|>
  82. [82]
    Leica LMD Systems Laser Microdissection Microscopes | Products
    Leica LMD systems allow users to easily isolate Regions of Interest (ROI) from entire areas of tissue down to single cells or even subcellular structures such ...Applications · Tech Specs · Publications · Downloads
  83. [83]
    Leica LMD6500 & LMD7000 Laser Microdissection Systems | Products
    Free deliveryLaser Microdissection (LMD) makes it possible to distinguish between relevant and non-relevant cells or tissues.
  84. [84]
    [PDF] Leica LMD6/7
    The Leica LMD systems perform sample preparation for molecular biology analysis directly from the tissue section using a UV laser.
  85. [85]
    Leica LMD6 & LMD7 - Opti-Tech Scientific Inc
    Aug 2, 2023 · It features the Pixel Classifier tool, which automatically detects and defines Regions of Interest (ROI) for Laser Microdissection (LMD). Aivia ...
  86. [86]
    Leica LMD Systems Laser Microdissection Microscopes - Applications
    Laser Microdissection (LMD, also known as Laser Capture Microdissection or LCM) enables users to isolate specific single cells or entire areas of tissue.Dissection Perfection · Biomarker Discovery With... · Molecular Biology Analysis...
  87. [87]
    How did Laser Microdissection enable Pioneering Neuroscience ...
    Jun 24, 2024 · LMD enabled single-cell analysis, captured daughter cells, and was used to trace cellular lineage, advancing understanding of adult ...
  88. [88]
    About MMI | Innovation in Single-Cell Tech - Molecular Machines
    Molecular Machines & Industries (MMI) is the leading provider of microscope ... Founded in 1992 by Prof. Dr. Stefan Seeger in Heidelberg, Germany, MMI ...
  89. [89]
    Molecular Machines & Industries (MMI) and ZEISS Announce ...
    Effective immediately, MMI and ZEISS will join forces to provide cutting-edge solutions for isolating individual cells, subcellular compartments ...
  90. [90]
    [PDF] Isolate single cells. - Zeiss
    MMI CellCut uses gentle laser microdissection for precise, contamination-free isolation of single cells, even living cells, from various tissues.Missing: MM300 CS100
  91. [91]
    [PDF] Solutions for - Molecular Pathology
    MMI has developed a laser microdissection technology with unmatched precision and speed where sample isolation is absolute contamination free.Missing: qPCR | Show results with:qPCR
  92. [92]
    MMI attended Lab-on-a-Chip and Microfluidics Europe 2025
    MMI attended Lab-on-a-Chip and Microfluidics Europe 2025 - MMI showcased how the CellManipulator can enhance Lab-on-a-Chip functionality.Missing: LCM 2024
  93. [93]
    Microdissection—An Essential Prerequisite for Spatial Cancer Omics
    Jun 23, 2020 · According to publications listed in pubmed, microdissection reached a plateau with 70–80 studies in 2007. In the same year we speculated in our ...<|control11|><|separator|>
  94. [94]
    Cell Isolation Solutions - Molecular Machines
    1. The most gentle isolation. LCM with the MMI CellCut and ZEISS Axio Observer features the cleanest laser on the market for dissection. · 2. Check your ...Missing: Leica LMD
  95. [95]
    [PDF] MMI CellCut - Cellmanipulator A4 - Molecular Machines
    • Repositioning accuracy: < 1 µm. • Step resolution: 0.156 µm. • Speed: 50 mm/sec. Long travel scanning stage. • Scanning area: 280 × 82 mm2. • Repositioning ...
  96. [96]
    [PDF] Technical Information - Light Microscopy Core Facility
    PALM MicroBeam works with a pulsed frequency tripled solid state laser (wavelength: 355 nm) which is coupled via the fluorescence path into a research ...
  97. [97]
    Laser Capture Microdissection Market | Industry Report, 2030
    The global laser capture microdissection market size was valued at USD 203.1 million in 2024 and is projected to reach USD 329.0 million by 2030, ...
  98. [98]
    Review of Laser Microdissection Systems | Biocompare
    Aug 26, 2021 · Laser microdissection systems allow you to dissect a tiny region of interest—such as a cell cluster, single cell, or nucleus—from sample tissue ...