LMX1B Human

LIM Homeobox Transcription Factor 1, Beta Human Recombinant
Shipped with Ice Packs
In Stock

Description

Limb Patterning

LMX1B establishes dorsoventral polarity by suppressing ventral structures and promoting dorsal autopod (hand/foot) and zeugopod (forearm/leg) development. Loss-of-function mutations disrupt this axis, causing NPS-associated skeletal abnormalities.

Developmental RoleMechanismSource
Dorsal Limb MesenchymeActivates enhancers (e.g., LARM1/LARM2) to amplify LMX1B expression
Nail and Patella FormationRegulates dorsal-specific genes; mutations lead to hypoplasia/aplasia

Kidney and Eye Development

LMX1B governs glomerular basement membrane integrity and anterior eye segment formation. In Lmx1b knockout mice, glomerulopathy and cataracts occur due to disrupted podocyte differentiation and collagen synthesis.

Tissue/OrganFunctionSource
KidneysRegulates COL4A3/A4, PODOCIN (podocyte structure)
EyesRequired for anterior segment morphogenesis (iris, cornea)
Nervous SystemMaintains dopaminergic (mDANs) and serotonergic neurons

Clinical Relevance: Nail-Patella Syndrome (NPS)

NPS is caused by haploinsufficiency of LMX1B due to mutations or deletions. Key features include:

  • Skeletal: Hypoplastic nails, absent/knee patellae, elbow dislocation.

  • Renal: Glomerular basement membrane defects, chronic nephropathy.

  • Ocular: Glaucoma, cataracts.

Genetic Causes of NPS

Mutation TypeFunctional ImpactExample MutationsSource
Coding RegionMissense, nonsense, frameshift; disrupt DNA bindingR198X, Δ246Ν247Q, V242L
Gene DeletionsWhole/partial loss of LMX1B expressionExon 2–5 deletions
5'UTR VariantsIntroduce uORFs, trigger NMD; reduced protein-174C>T, -226G>A

Mechanism of Pathogenicity:

  • Loss-of-function: Mutations impair LMX1B’s ability to bind DNA or recruit coactivators (e.g., E47/shPan1) .

  • Dominant-negative effect: Absent in most cases; haploinsufficiency drives NPS .

LMX1B and Autophagy: Emerging Interactions

Recent studies reveal LMX1B’s role in mitochondrial quality control and autophagy in neurons:

ProcessLMX1B’s RoleSource
Autophagy RegulationBinds ATG8 proteins (LC3B); activates ULK1, ATG7, TFEB
Mitochondrial ROSOverexpression reduces ROS; suppression elevates oxidative stress
NeuroprotectionInduces autophagy flux; protects against rotenone toxicity in mDANs

Mechanistic Insight:

  • ATG8 Binding: LMX1B binds LC3B in the nucleus under basal conditions, enhancing transcription of autophagy genes.

  • Mitochondrial Respiration: Suppression reduces oxidative phosphorylation, increasing vulnerability to stressors.

Functional Characterization of Mutants

MutationDNA BindingTransactivation ActivityDominant-Negative EffectSource
Δ246Ν247QLost~30% of wild-typeNone
V242LReduced~50% of wild-typeNone
5'UTR uORF VariantsN/A~20% of wild-typeNone

Key Findings:

  • Homeodomain Mutations: Critical for DNA binding; substitutions (e.g., R223X) disrupt helix-loop-helix interactions .

  • uORF Variants: 5'UTR mutations (e.g., -174C>T) introduce upstream ORFs, reducing LMX1B translation via leaky ribosome scanning .

Regulatory Elements

  • LARM1/LARM2 Enhancers: Auto-regulatory modules amplifying LMX1B expression in limbs; deletions cause NPS with restricted limb/nail phenotypes .

  • 3D Chromatin Interactions: LARM regions physically interact with the LMX1B promoter, enabling feedback loops .

Therapeutic Potential

  • Gene Therapy: Restoration of LMX1B expression in NPS models.

  • Autophagy Modulation: Targeting LMX1B-ATG8 interactions to protect neurons in Parkinson’s disease .

References

  1. ATG8-dependent LMX1B-autophagy crosstalk

  2. Functional Characterization of LMX1B Mutations

  3. LMX1B Gene Overview

  4. Entire LMX1B Deletions in NPS

  5. LMX1B Transcriptional Regulation

  6. Limb-Specific Lmx1b Modules

  7. 5'UTR Variants in NPS

Product Specs

Introduction
LIM Homeobox Transcription Factor 1, Beta (LMX1B) belongs to the LIM-homeodomain protein family. These proteins are characterized by two N-terminal zinc-binding LIM domains, a homeodomain, and a C-terminal glutamine-rich domain. Functioning as a transcription factor, LMX1B is crucial for determining dorsal limb fate in the zeugopodal and autopodal stages of development. Mutations in the LMX1B gene have been linked to nailpatella syndrome.
Description
Recombinant human LMX1B protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 418 amino acids (1-395a.a), including a 23 amino acid His-tag at the N-terminus, and has a molecular weight of 46.5kDa. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless, and sterile-filtered solution.
Formulation
The LMX1B protein is supplied in a solution at a concentration of 1mg/ml. The solution contains 20mM Tris-HCl buffer (pH 8.0), 10% glycerol, and 0.4M Urea.
Stability
For short-term storage (up to 4 weeks), the protein can be stored at 4°C. For extended periods, it is recommended to store the protein frozen at -20°C. To ensure long-term stability, adding a carrier protein (0.1% HSA or BSA) is advisable. Repeated freezing and thawing of the protein should be avoided.
Purity
The purity of the LMX1B protein is greater than 80%, as determined by SDS-PAGE analysis.
Synonyms
LIM homeobox transcription factor 1, beta, LMX1.2, NPS1, LIM/homeobox protein 1.2, LIM/homeobox protein LMX1B, LMX1B.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMDIATGP ESLERCFPRG QTDCAKMLDG IKMEEHALRP GPATLGVLLG SDCPHPAVCE GCQRPISDRF LMRVNESSWH EECLQCAACQ QALTTSCYFR DRKLYCKQDY QQLFAAKCSG CMEKIAPTEF VMRALECVYH LGCFCCCVCE RQLRKGDEFV LKEGQLLCKG DYEKEKDLLS SVSPDESDSV KSEDEDGDMK PAKGQGSQSK GSGDDGKDPR RPKRPRTILT TQQRRAFKAS FEVSSKPCRK VRETLAAETG LSVRVVQVWF QNQRAKMKKL ARRHQQQQEQ QNSQRLGQEV LSSRMEGMMA SYTPLAPPQQ QIVAMEQSPY GSSDPFQQGL TPPQMPGNDS IFHDIDSDTS LTSLSDCFLG SSDVGSLQAR VGNPIDRLYS MQSSYFAS.

Q&A

What is LMX1B and what molecular domains characterize its function?

LMX1B is a LIM homeodomain transcription factor that emerged as a distinct paralog alongside the development of complex chordate brain architecture . It contains highly conserved functional domains including a homeodomain (100% homology with LMX1A) and LIM domains (LIMA [67% homology] and LIMB [83% homology] compared to LMX1A) . These domains are critical for DNA binding and protein-protein interactions that mediate its transcriptional activities. LMX1B binds to A/T-rich FLAT elements in gene promoters to regulate transcription of target genes involved in development, autophagy, and cellular stress responses . Unlike LMX1A, which primarily regulates neurogenesis in the ventral mesencephalic floor plate and inner ear development, LMX1B has broader roles in brain development, particularly in dorsoventral patterning and serotonergic neuron development, as well as in coordinating aspects of eye, limb, and kidney development .

How does LMX1B regulate autophagy and stress protection in human cells?

LMX1B functions as an autophagy transcription factor that provides cellular stress protection through multiple mechanisms:

  • Transcriptional regulation: LMX1B binds to FLAT elements in the promoters of key autophagy genes including ULK1, ATG3, ATG16L1, UVRAG, TFEB, NDP52, OPTN, and PINK1 .

  • Protein interactions: LMX1B binds to multiple ATG8 family proteins, with these interactions being dependent on a conserved region C-terminal to the homeodomain .

  • Subcellular activity: LMX1B interacts with LC3B (an ATG8 protein) in the nucleus under basal conditions but associates with both cytosolic and nuclear LC3B during nutrient starvation .

  • Stress protection: LMX1B suppression dampens autophagy responses, lowers mitochondrial respiration, and elevates mitochondrial reactive oxygen species (ROS) . Conversely, inducible overexpression of LMX1B protects against rotenone toxicity in human iPSC-derived midbrain dopaminergic neurons in vitro .

These findings establish a novel LMX1B-autophagy regulatory axis that contributes to mDAN maintenance and survival in the adult brain .

What experimental models are currently used to study LMX1B function?

Researchers employ several experimental models to study LMX1B function:

  • Knockout mouse models:

    • Systemic Lmx1b knockdown mice exhibit skeletal and kidney defects consistent with Nail-Patella syndrome pathology, leading to neonatal lethality approximately 24 hours after birth .

    • Conditional knockout models using Cre-loxP systems (e.g., Pet1-Cre for targeting serotonergic neurons) allow for temporal and tissue-specific ablation of Lmx1b .

  • Human cell lines and iPSC models:

    • Human induced pluripotent stem cell (iPSC)-derived midbrain dopaminergic neurons provide a platform for studying LMX1B's role in human neuronal development and protection .

    • These models allow for inducible overexpression or suppression of LMX1B to assess its functions .

  • Lineage tracing systems:

    • TdTomato reporter systems have been used to visualize and track LMX1B-expressing neurons and their projections .

  • Temporal conditional targeting approaches:

    • Stage-specific targeting allows researchers to dissect distinct functions of LMX1B at successive developmental stages .

These models collectively enable researchers to examine LMX1B's roles in development, transcriptional regulation, and disease pathogenesis.

What methodologies can be used to investigate the LMX1B-ATG8 protein interaction dynamics?

To investigate LMX1B-ATG8 protein interaction dynamics, researchers can employ several methodological approaches:

  • Co-immunoprecipitation (Co-IP): This technique has revealed that LMX1B interacts with LC3B (an ATG8 protein) in different subcellular compartments depending on nutrient status .

  • Subcellular fractionation: This approach helps determine the localization of LMX1B-ATG8 interactions, showing that LMX1B interacts with LC3B in the nucleus under basal conditions but associates with both cytosolic and nuclear LC3B during nutrient starvation .

  • Domain mapping: Mutational analysis of the conserved region C-terminal to the homeodomain of LMX1B can identify specific residues required for ATG8 binding .

  • Nutrient deprivation experiments: Starvation conditions can be used to study how nutrient status affects LMX1B-ATG8 interactions and subcellular localization .

  • Transcriptional reporter assays: These assays determine how ATG8 binding influences LMX1B's transcriptional activity, showing that ATG8 binding stimulates LMX1B-mediated transcription for efficient autophagy and cell stress protection .

  • Fluorescence microscopy: This technique visualizes the co-localization of LMX1B with ATG8 proteins in different cellular compartments under various conditions .

These approaches collectively provide insights into the complex dynamics of LMX1B-ATG8 interactions and their functional consequences.

How does LMX1B regulate stage-specific development of serotonergic axon projections?

LMX1B regulates serotonergic axon development through stage-specific mechanisms:

  • Primary outgrowth stage: LMX1B controls the initial growth rate of primary 5-HT axon pathways .

  • Pathway routing stage: LMX1B mediates selective routing of serotonergic axons to ensure proper connectivity .

  • Terminal arborization stage: LMX1B controls the final branching and arborization of 5-HT axons through an LMX1B→Pet1 regulatory cascade .

  • Molecular regulation: LMX1B upregulates expression of key axon arborization genes, such as Protocadherin-alphac2, during postnatal development of forebrain 5-HT axons .

  • Distinct pathways: LMX1B differentially regulates ascending and descending 5-HT pathways, with an ascending-specific regulatory cascade controlling forebrain 5-HT axon arborization .

Research using conditional knockout mouse models has demonstrated that loss of LMX1B results in failure to generate axonal projections to the forebrain and spinal cord . Experimental approaches using TdTomato reporter systems have shown that although TdTomato-positive axons are present in normal density and with proper ascending trajectory within the medial forebrain bundle (MFB) of Lmx1b conditional knockout mice, these axons are nearly completely missing throughout the forebrain .

What experimental evidence supports LMX1B's role in Parkinson's disease pathophysiology?

Several lines of experimental evidence support LMX1B's role in Parkinson's disease (PD) pathophysiology:

  • Expression correlation: Patient brain LMX1B levels have been reported to inversely correlate with PD progression, suggesting a protective role .

  • Genetic associations: LMX1B polymorphisms have been linked (albeit weakly) to PD susceptibility .

  • Conditional knockout mouse models: Targeted Lmx1a/Lmx1b ablation in mice triggers midbrain dopaminergic neuron decline associated with neuropathological features (e.g., α-synuclein–positive, distended axonal terminals) and behavioral abnormalities consistent with PD .

  • Age-related expression: Lmx1a expression declines with age in the mouse brain, potentially explaining increased vulnerability to PD with aging .

  • Neuroprotective mechanisms: LMX1B overexpression protects against rotenone toxicity (a PD model) in human iPSC-derived midbrain dopaminergic neurons in vitro .

  • Cellular stress resistance: LMX1B provides cellular stress protection through promoting autophagy, maintaining mitochondrial respiration, and reducing mitochondrial ROS—all processes implicated in PD pathogenesis .

These findings collectively suggest that maintaining LMX1B in the adult brain is important for protection against PD-associated midbrain dopaminergic neuron decline, while boosting expression and/or activities of this transcription factor may offer therapeutic benefits .

How can researchers experimentally distinguish between direct and indirect transcriptional targets of LMX1B?

Distinguishing between direct and indirect transcriptional targets of LMX1B requires a multi-faceted experimental approach:

  • Bioinformatic analysis of promoter regions: Researchers can search for potential LMX1B-targeting A/T-rich FLAT elements in gene promoter regions . This approach has identified FLAT elements in known LMX1B target genes like COL4A3, IFNB1, NURR1, PITX3, and TH, as well as in autophagy-related genes including ULK1, ATG3, ATG16L1, and PINK1 .

  • Chromatin immunoprecipitation (ChIP): This technique identifies direct DNA binding sites of LMX1B by immunoprecipitating LMX1B-bound chromatin fragments and sequencing the associated DNA.

  • Reporter gene assays: By cloning promoter regions of potential target genes upstream of reporter genes, researchers can assess whether LMX1B directly activates transcription from these promoters.

  • Time-course expression analysis: Following LMX1B induction or suppression, early-response genes are more likely to be direct targets, while later-response genes may be indirect targets.

  • Combinatorial approaches with ATG8 binding: Since ATG8 proteins stimulate LMX1B-mediated transcription, comparing transcription patterns with and without disruption of ATG8-LMX1B interaction can help identify direct targets dependent on this regulatory mechanism .

These methodologies collectively allow researchers to build confidence in identifying direct versus indirect transcriptional targets of LMX1B.

What is the molecular basis for the differential functions of LMX1A and LMX1B despite their high sequence homology?

The differential functions of LMX1A and LMX1B despite their high sequence homology (100% in homeodomain, 67% in LIMA, and 83% in LIMB) can be attributed to several molecular mechanisms:

  • Distinct tissue expression patterns: LMX1A and LMX1B have different spatial and temporal expression patterns despite their sequence similarity .

  • Protein interaction partners: While both factors bind ATG8 proteins, they may interact with different tissue-specific cofactors that direct their activity to different genomic loci or modify their function.

  • Regulatory elements: Different enhancers and repressors may control LMX1A versus LMX1B expression in different tissues, leading to their distinct roles .

  • Functional compensation: Despite their differences, Lmx1a and Lmx1b can partially compensate for one another with respect to midbrain dopaminergic neuron specification in mice , suggesting overlapping functions in certain contexts.

  • Evolutionary specialization: LMX1A and LMX1B emerged as distinct paralogs alongside the development of more complex chordate brain architecture, suggesting evolutionary specialization of their functions .

What approaches can be used to study the impact of LMX1B on mitochondrial function and oxidative stress?

To study LMX1B's impact on mitochondrial function and oxidative stress, researchers can employ several methodological approaches:

  • Mitochondrial respiration assays: Oxygen consumption rate (OCR) measurements using platforms like Seahorse XF Analyzer can assess how LMX1B affects mitochondrial respiratory capacity .

  • ROS detection assays: Fluorescent probes specific for mitochondrial ROS (e.g., MitoSOX) can measure how LMX1B suppression elevates mitochondrial ROS levels .

  • Live-cell imaging of mitochondrial dynamics: Fluorescent markers for mitochondria can visualize changes in mitochondrial morphology, distribution, and function following LMX1B manipulation.

  • Mitophagy assessment: Dual-fluorescence reporters can track mitochondrial clearance through autophagy (mitophagy) in relation to LMX1B activity.

  • Neurotoxin models: Exposing neurons to mitochondrial toxins like rotenone (which inhibits complex I) can assess how LMX1B overexpression protects against mitochondrial dysfunction in human iPSC-derived midbrain dopaminergic neurons .

  • Transcriptomic analysis: RNA-seq following LMX1B manipulation can identify changes in expression of genes involved in mitochondrial function, oxidative stress response, and related pathways.

These approaches collectively provide a comprehensive assessment of how LMX1B influences mitochondrial health and oxidative stress resistance in neurons.

What experimental systems best model LMX1B mutations associated with Nail-Patella syndrome?

Researchers studying LMX1B mutations associated with Nail-Patella syndrome can utilize several experimental systems:

  • Patient-derived iPSCs: Generating induced pluripotent stem cells from Nail-Patella syndrome patients allows for differentiation into relevant cell types (podocytes, chondrocytes, neurons) to study disease mechanisms.

  • CRISPR/Cas9 gene editing: Introducing specific LMX1B mutations in the LIM or homeodomain regions allows researchers to model Nail-Patella syndrome in cellular or animal models .

  • Conditional knockout models: Tissue-specific knockout of Lmx1b in podocytes triggers proteinuria linked to podocyte actin disorganization and slit diaphragm failure, mimicking Nail-Patella syndrome kidney manifestations .

  • Systemic knockout models: Systemic Lmx1b knockdown in mice produces skeletal and kidney defects consistent with Nail-Patella syndrome pathology .

  • Domain-specific functional assays: Testing how mutations in different LMX1B domains affect:

    • DNA binding capacity

    • Protein-protein interactions (especially with ATG8 proteins)

    • Transcriptional activity at target genes like COL4A3

    • Cellular localization and stability

These models help researchers understand how LMX1B mutations lead to the tissue-specific manifestations of Nail-Patella syndrome, including skeletal developmental abnormalities, chronic nephropathy, and open-angle glaucoma .

How can temporal targeting approaches reveal stage-specific functions of LMX1B?

Temporal targeting approaches are critical for dissecting LMX1B's stage-specific functions:

  • Conditional genetic systems: Using inducible Cre-loxP systems (e.g., with tamoxifen-inducible promoters) allows for temporal control of Lmx1b ablation at different developmental stages .

  • Viral-mediated gene delivery: Stereotaxic injection of viral vectors expressing Cre recombinase at specific developmental timepoints enables temporal control of Lmx1b knockout in targeted brain regions.

  • Sequential analysis: Examining the consequences of LMX1B loss at successive stages of neurodevelopment has revealed its distinct roles in primary pathway growth rate, selective pathway routing, and terminal arborization of serotonergic axons .

  • Lineage tracing with stage-specific reporters: Combining temporal LMX1B manipulation with reporter systems (like TdTomato) allows visualization of stage-specific consequences on neuronal development and projection patterns .

  • Transcriptional profiling at distinct stages: RNA-seq analysis following stage-specific LMX1B manipulation identifies temporally regulated gene networks.

Research using these approaches has demonstrated that a single continuously expressed transcription factor (LMX1B) can act at successive stages to build expansive axon pathway architectures, enabling CNS-wide serotonergic neuromodulation .

What therapeutic strategies might target the LMX1B pathway for neurodegenerative diseases?

Several therapeutic strategies targeting the LMX1B pathway show promise for neurodegenerative diseases:

  • Gene therapy approaches: Delivering functional LMX1B to midbrain dopaminergic neurons could potentially slow neurodegeneration in Parkinson's disease, given that LMX1B inducible overexpression protects against rotenone toxicity in human iPSC-derived midbrain dopaminergic neurons .

  • Small molecule enhancers: Developing compounds that enhance LMX1B transcriptional activity or stability could boost its neuroprotective effects. Research shows that LMX1B levels inversely correlate with PD progression, suggesting therapeutic potential in maintaining or increasing LMX1B levels .

  • ATG8-LMX1B interaction modulators: Since ATG8 binding stimulates LMX1B-mediated transcription for efficient autophagy and cell stress protection, compounds that enhance this interaction could boost neuroprotection .

  • Targeting downstream pathways: Modulating specific downstream targets of LMX1B involved in autophagy (e.g., ULK1, ATG3, ATG16L1) or mitochondrial function could provide alternative therapeutic avenues .

  • Combined approaches: Strategies that simultaneously enhance LMX1B levels/activity while boosting autophagy through complementary mechanisms might provide synergistic benefits for neuroprotection.

These approaches are supported by findings that LMX1B acts as an autophagy transcription factor providing cellular stress protection, with its suppression dampening autophagy responses, lowering mitochondrial respiration, and elevating mitochondrial ROS .

What challenges exist in translating LMX1B research from animal models to human applications?

Translating LMX1B research from animal models to human applications faces several significant challenges:

  • Species-specific differences: Despite high conservation, there may be species-specific differences in LMX1B function, regulation, or downstream targets between mice and humans.

  • Cell-type specificity: LMX1B functions differently across tissues (midbrain, serotonergic neurons, kidney, limbs), necessitating precise targeting to avoid off-target effects in therapeutic applications .

  • Developmental timing: LMX1B has distinct stage-specific functions during development and adulthood, requiring careful consideration of timing for therapeutic interventions .

  • Technical delivery challenges: Delivering LMX1B modulators specifically to affected neural populations (e.g., midbrain dopaminergic neurons) remains technically challenging.

  • Regulatory complexity: The LMX1B-autophagy regulatory axis involves complex interactions with ATG8 proteins and other factors that may differ between experimental models and human patients .

  • Disease heterogeneity: Conditions like Parkinson's disease involve multiple pathogenic mechanisms beyond LMX1B dysregulation, potentially limiting the effectiveness of LMX1B-targeted approaches in diverse patient populations.

Addressing these challenges requires continued research using human cell models (such as iPSC-derived neurons) alongside traditional animal models to better understand LMX1B biology in the human context .

What are the most promising research frontiers in understanding LMX1B function?

Several promising research frontiers are emerging in LMX1B research:

  • Single-cell transcriptomics: Applying single-cell RNA-seq to LMX1B-expressing tissues will reveal cell-specific roles and regulatory networks across development and disease states.

  • Structural biology: Determining the three-dimensional structure of LMX1B in complex with ATG8 proteins would provide mechanistic insights into how these interactions stimulate transcriptional activity .

  • Long-term in vivo imaging: Developing techniques to visualize LMX1B-dependent neuronal projections over time in living organisms could reveal dynamic aspects of its function in circuit formation and maintenance .

  • Systems biology approaches: Integrating transcriptomic, proteomic, and metabolomic data following LMX1B manipulation would provide a comprehensive understanding of its effects on cellular physiology.

  • Translational research: Moving from basic mechanisms to therapeutic applications by testing LMX1B pathway modulators in patient-derived cells and preclinical models of Parkinson's disease and other LMX1B-associated conditions .

These research directions hold promise for advancing our understanding of LMX1B biology and developing novel therapeutic approaches for LMX1B-related disorders.

How might advances in gene editing technologies influence LMX1B research?

Advances in gene editing technologies are transforming LMX1B research in several ways:

  • Precise mutation modeling: CRISPR/Cas9 enables precise introduction of patient-specific LMX1B mutations to study their functional consequences in cellular and animal models.

  • Conditional expression systems: Advanced gene editing tools allow for the development of sophisticated inducible systems to control LMX1B expression with unprecedented temporal and spatial precision .

  • Domain-specific modifications: Targeted modifications of specific LMX1B domains can dissect the functional importance of different regions, such as the conserved region C-terminal to the homeodomain that mediates ATG8 binding .

  • High-throughput screening: CRISPR libraries targeting LMX1B pathway components can identify novel regulatory factors and potential therapeutic targets.

  • In vivo gene therapy approaches: Advancements in delivery methods for gene editing tools may eventually enable direct correction of LMX1B mutations or modulation of LMX1B expression in patients with related disorders.

Product Science Overview

Introduction

LIM Homeobox Transcription Factor 1, Beta (LMX1B) is a protein encoded by the LMX1B gene in humans. This transcription factor is a member of the LIM-homeodomain family of proteins, which are characterized by the presence of two N-terminal zinc-binding LIM domains, a homeodomain, and a C-terminal glutamine-rich domain .

Gene and Protein Structure

The LMX1B gene is located on chromosome 9 and encodes a protein consisting of 402 amino acids . The protein structure includes:

  • Two N-terminal LIM domains: These domains are involved in protein-protein interactions and are crucial for the transcriptional activity of LMX1B.
  • Homeodomain: This domain is responsible for DNA binding and is essential for the transcriptional regulation of target genes.
  • C-terminal glutamine-rich domain: This domain is involved in transcriptional activation .
Biological Functions

LMX1B plays a critical role in various developmental processes, including:

  • Dorsal limb development: LMX1B is essential for the specification of dorsal limb structures at both the zeugopodal (forearm and lower leg) and autopodal (hand and foot) levels .
  • Renal glomerular filtration barrier: It is involved in the regulation of podocyte-expressed genes, which are crucial for the integrity of the glomerular basement membrane .
  • Anterior segment of the eye: LMX1B is necessary for the normal development of the anterior segment of the eye .
  • Neuronal differentiation: It plays a role in the differentiation of dopaminergic and serotonergic neurons .
Clinical Significance

Mutations in the LMX1B gene are associated with several disorders, including:

  • Nail-Patella Syndrome (NPS): This genetic disorder is characterized by abnormalities in the nails, knees, elbows, and pelvis. It is caused by mutations in the LMX1B gene .
  • Focal Segmental Glomerulosclerosis 10 (FSGS10): This condition affects the kidneys and is also linked to mutations in LMX1B .
Recombinant LMX1B

Human recombinant LMX1B is produced using recombinant DNA technology, which involves inserting the LMX1B gene into a suitable expression system, such as bacteria or mammalian cells. This allows for the production of large quantities of the protein for research and therapeutic purposes.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.