bims-mecosi Biomed News
on Membrane contact sites
Issue of 2026–01–04
thirteen papers selected by
Verena Kohler, Umeå University



  1. Prog Lipid Res. 2025 Dec 26. pii: S0163-7827(25)00054-2. [Epub ahead of print]101 101372
      Membrane contact sites (MCSs) are fundamental hubs of inter-organelle communication that mediate the non-vesicular exchange of lipids, ions, and metabolites, thereby sustaining cellular homeostasis. In plants, the "contactome"-the dynamic network of all membrane contact sites-has evolved distinctive features to accommodate the requirements of a sessile, photosynthetic lifestyle and the presence of plastids. Within this network, the endoplasmic reticulum (ER) functions as a central hub for lipid biosynthesis and distribution, forming functionally important contacts with multiple organelles. Recent advances in high-resolution imaging, lipidomics, and molecular genetics are beginning to uncover the complexity of these inter-organelle connections and their contribution to lipid homeostasis in plants. This review summarizes current knowledge of the plant contactome, with a focus on lipid transfer proteins and lipid-modifying enzymes that maintain lipid balance during organelle biogenesis, plant development, and stress adaptation. Plant lipid transfer at membrane contact sites can be broadly divided into two mechanistic modes: precision-regulated "shuttles," exemplified by the Ca2+-dependent SYT1-mediated diacylglycerol transfer at ER-plasma membrane interfaces, and high-capacity lipid transfer mechanisms, such those mediated by ATG2, that support rapid lipid flux during autophagosome biogenesis. Knowledge of lipid metabolism at plant membrane contact sites is still in its initial stages, and many of the underlying mechanisms remain unexplored. Major challenges include understanding how these sites integrate stress responses, metabolic fluxes, and organelle dynamics. Addressing these questions will be essential to unravel the unique aspects of plant lipid biology and may open opportunities for improving stress resilience and metabolic engineering in crops.
    Keywords:  Contactome; Endoplasmic reticulum; Lipid metabolism; Lipid transfer proteins; Membrane contact sites; Non-vesicular transport; Organelle communication
    DOI:  https://doi.org/10.1016/j.plipres.2025.101372
  2. CNS Neurosci Ther. 2025 Dec;31(12): e70726
       BACKGROUND: Mitochondria-associated endoplasmic reticulum membranes (MAMs) are specialized regions in cells where the endoplasmic reticulum and mitochondria closely interact. MAMs are enriched with a variety of proteins that regulate key cellular processes. These processes include mitochondrial fission and fusion, autophagy, lipid metabolism, calcium homeostasis, and oxidative stress. Increasing evidence suggests that disruption of MAMs structure and alterations in associated protein expression patterns are closely related to the pathogenesis of epilepsy.
    METHODS: This review synthesizes and analyzes current literature to outline the structural and functional roles of key MAMs proteins. It further examines experimental and clinical evidence linking MAMs dysregulation to epileptogenesis and treatment responses.
    RESULTS: The analysis confirms that MAMs serve as a central hub coordinating cellular homeostasis. Specific alterations in MAMs structure and protein expression are consistently associated with epilepsy models. These alterations directly impact neuronal excitability, synaptic function, and cell survival pathways involved in disease progression.
    CONCLUSION: Addressing these structural and functional properties of MAMs may provide valuable insights for developing novel therapeutic strategies for epilepsy.
    Keywords:  endoplasmic reticulum; epilepsy; mitochondria; mitochondria‐associated endoplasmic reticulum membranes; therapeutic targets
    DOI:  https://doi.org/10.1002/cns.70726
  3. Neural Regen Res. 2025 Dec 30.
      Mitochondria-associated endoplasmic reticulum membranes serve as crucial signaling hubs mediating communication between the endoplasmic reticulum and mitochondria, and play a central role in calcium ion exchange. This dynamic interface regulates key cellular processes including bioenergetic metabolism, apoptosis, autophagy, and stress responses. Dysregulation of calcium transport associated with mitochondria-associated endoplasmic reticulum membranes can disrupt intracellular homeostasis, leading to mitochondrial dysfunction, oxidative stress, and neuronal death, which are hallmarks of aging and neurodegenerative diseases. This review systematically examines the functions of protein complexes within mitochondria-associated endoplasmic reticulum membranes and the pathogenic mechanisms of calcium signaling regulated by these membranes in neurodegenerative disorders. It places particular emphasis on structural alterations in calcium ion transport machinery as a common mechanism underlying various neurodegenerative diseases. In Alzheimer's disease, mitochondria-associated endoplasmic reticulum membranes exhibit a hyperactive state, promoting the generation of amyloid-β and enhancing calcium ion flux from the endoplasmic reticulum to the mitochondria. In contrast, in Parkinson's disease and amyotrophic lateral sclerosis, the activity of mitochondria-associated endoplasmic reticulum membranes is reduced, leading to a decline in mitochondrial calcium ion buffering capacity and exacerbating excitotoxicity. Proteins residing in mitochondria-associated endoplasmic reticulum membranes are disrupted across various neurodegenerative diseases, resulting in abnormal communication between the endoplasmic reticulum and mitochondria. Recent studies indicate that mitochondria-associated endoplasmic reticulum membranes play a bidirectional role in disease progression, and compensatory mechanisms often exacerbate the pathological process. Therapeutic strategies aimed at preserving the integrity of mitochondria-associated endoplasmic reticulum membranes hold promise for alleviating neurodegenerative damage. Therefore, calcium ion exchange mediated by mitochondria-associated endoplasmic reticulum membranes plays a key role in aging and neurodegenerative diseases, making it a highly promising therapeutic target.
    Keywords:  Alzheimer's disease; Parkinson's disease; aging; amyotrophic lateral sclerosis; calcium channels; frontotemporal dementia; mitochondria-associated endoplasmic reticulum membranes; multiple sclerosis; neurodegenerative diseases; programmed cell death
    DOI:  https://doi.org/10.4103/NRR.NRR-D-25-00857
  4. Front Cardiovasc Med. 2025 ;12 1751239
      
    Keywords:  endoplasmic reticulum; heart; heart-related diseases; mitochondria; mitochondria-associated endoplasmic reticulum membranes
    DOI:  https://doi.org/10.3389/fcvm.2025.1751239
  5. Neurobiol Dis. 2025 Dec 25. pii: S0969-9961(25)00465-6. [Epub ahead of print] 107248
      Although targeting neuronal excitability remains the cornerstone of epilepsy treatment, the high prevalence of drug-resistant epilepsy compels a reexamination of its upstream mechanisms. Growing evidence identifies redox imbalance and specific cell death programs as key drivers of epileptogenesis. We propose a unified framework. Here, we position dysfunction of mitochondria-associated endoplasmic reticulum membranes (MAMs) linked to ferroptosis as a core pathogenic axis. Multiple epileptogenic triggers converge to pathologically remodel MAMs, transforming them into a catalytic platform that efficiently initiates ferroptosis. This is achieved through the nanoscale co-localization of calcium ions, reactive oxygen species, and unstable iron. We systematically dissect how MAMs integrate calcium signaling, lipid metabolism, and redox balance, and outline core ferroptosis pathways. Critically, MAMs remodeling subverts antioxidant defenses, reprograms lipid metabolism, and irreversibly drives ferroptosis. This MAMs-ferroptosis axis promotes epilepsy chronicity by mediating selective neuronal loss, amplifying neuroinflammation, and disrupting excitatory-inhibitory balance. Based on this mechanism, we propose a novel therapeutic paradigm: stabilizing MAMs upstream with Sigma-1 receptor ligands, combined with neutralizing lipid peroxides downstream using ferroptosis inhibitors. This multi-tiered strategy provides a foundation for developing disease-modifying, next-generation epilepsy therapies.
    Keywords:  Epilepsy; Ferroptosis; Lipid peroxidation; Mitochondrial-associated endoplasmic reticulum membranes; Reactive oxygen species; Sigma-1 receptor
    DOI:  https://doi.org/10.1016/j.nbd.2025.107248
  6. Animal Model Exp Med. 2025 Dec 30.
       BACKGROUND: Mastitis seriously affects the mammary health of humans and animals. Studies have found that inflammation and oxidative stress play key roles in the occurrence and development of mastitis. Therefore, in-depth research on related molecular mechanisms is of great significance.
    METHODS: Postpartum mice were anesthetized with pentobarbital and administered lipopolysaccharide to develop the mouse mastitis model. Proteomic analysis was performed to compare protein expression in mitochondria-associated endoplasmic reticulum membranes (MAM) from two mouse mammary gland groups. Western blot was used to detect the expression of MAM-related proteins in mitochondria. AlphaFold3 was used to predict the molecular structures of phosphofurin acidic cluster sorting protein 2 (PACS2) and mitofusin 2 (MFN2) and their interaction levels. The MFN2-PACS2 interaction was investigated using co-immunoprecipitation and small interfering RNA.
    RESULTS: The results showed that the inflammation level in the mammary gland tissue of mice with mastitis significantly increased, the total antioxidant capacity decreased, and the expression of MAM-related proteins MFN2 and PACS2 was significantly downregulated. In cell experiments, overexpression of MFN2 can inhibit inflammation and oxidative stress responses, and promote the interaction between MFN2 and PACS2 to affect the formation of MAMs.
    CONCLUSION: In summary, this study suggests that mastitis can alter the expression of MAM-related proteins in mouse breast tissue. The interaction between MFN2 and PACS2 regulates the formation of MAMs. Overexpression of MFN2 can promote the formation of MAMs and inhibit inflammation and oxidative stress response in mammary epithelial cells. Our results provided a new theoretical basis and potential therapeutic targets for the prevention and treatment of mastitis.
    Keywords:  mastitis; mitochondria‐associated endoplasmic reticulum membranes (MAM); mitofusin 2 (MFN2); phosphofurin acidic cluster sorting protein 2 (PACS2)
    DOI:  https://doi.org/10.1002/ame2.70110
  7. J Endocr Soc. 2026 Jan;10(1): bvaf198
      Mitotane is an inhibitor of sterol O-acyltransferase 1 (SOAT1) approved for the treatment of adrenocortical carcinoma (ACC). In cells, mitotane increases reactive oxygen species, lipid peroxidation, and ultimately cell death. This mechanism is similar but distinct from ferroptosis, a cell death mechanism adrenal cortex cells are endogenously predisposed to. Both Acyl-CoA-Synthetase 4 (ACSL4), essential for ferroptosis, and SOAT1 are localized in mitochondria-associated membranes (MAM), specialized contact sites between mitochondria and endoplasmic reticulum (ER). Here, we used protein and lipid mass spectrometry to explore the role of MAMs in adrenocortical cell death. MAMs were isolated from NCI-H295S cells treated with mitotane, the ferroptosis inducer RSL3, or control. Western blotting of marker proteins was used for quality control prior to lipid and protein mass spectrometry. MAM fractions showed strong enrichment of SOAT1 and FATE1 (fetal and adult testis expressed 1) marker proteins, contained ACSL4, and were depleted from mitochondrial MTCO2 independent of treatment condition. Protein mass spectrometry identified IRE1alpha/ERN1, and PERK/EIF2AK3 implicated in the response to mitotane. Proteins involved in ER- and mitochondria-related processes were functionally enriched. We discovered the guanosine nucleotide exchange factor GRIPAP1 in MAMs of mitotane but not RSL3- or control-treated samples. In NCI-H295S cells mitotane upregulated GRIPAP1 expression. Mitotane but not RSL3 pronouncedly reduced the quantity of ubiquinone (Q10) and heme B in MAMs. In conclusion, locally reduced Q10 in MAM may contribute to impaired respiratory chain activity and free radical excess induced by mitotane. Recruitment of GRIPAP1 protein to MAMs may transduce cell death.
    Keywords:  adrenal gland; cancer treatment; cell death; ferroptosis; mitochondria-associated membranes; steroidogenesis
    DOI:  https://doi.org/10.1210/jendso/bvaf198
  8. Biochem Biophys Rep. 2026 Mar;45 102402
      Lipid droplets (LDs) maintain cellular lipid homeostasis through dynamic interactions with other organelles. Understanding how these contact sites form is crucial for uncovering the mechanisms of lipid exchange and signaling. In this study, we used an in vitro model to investigate how lipid composition and the LD-associated protein perilipin 5 (PLIN5) influence contact formation between an LD monolayer and a bilayer membrane. Artificial LDs consisting of triolein and coated with either a DOPE or DOPC monolayer containing PLIN5 or not were incubated with large unilamellar vesicles (LUVs) that mimic the bilayer membrane of the organelle. Using double fluorescence labeling of the LUV bilayer and the core, we can distinguish between fusion of the LUV bilayer with the LDs and stable attachment of LUVs to the LD's surface. Our results show that the probability of fusion between LDs and LUVs is greatly increased for DOPE-coated LDs, while PLIN5 promotes the stable attachment of LUVs to the LD's surface and prevents fusion. These observations illustrate how certain lipid and protein components can modulate contact formation between LDs and membranes in a controlled in vitro system, and provide a basis for future studies on the molecular mechanisms of organelle communication.
    Keywords:  Contact site; Large unilamellar vesicle; Lipid droplet; Lipidic bridge; PLIN5; Perilipin 5; Protein tether
    DOI:  https://doi.org/10.1016/j.bbrep.2025.102402
  9. Nat Chem Biol. 2026 Jan 02.
      Biopolymers that separate into condensed and dilute phases in solution also prewet membranes when one or more components couple to membrane lipids. Here we demonstrate that this prewetting transition becomes exquisitely sensitive to lipid composition when membranes have compositions near the boundary of liquid-ordered/liquid-disordered phase coexistence in both simulation and in reconstitution when polyelectrolytes are coupled to model membranes. In cells, we use an optogenetic tool to characterize prewetting at both the plasma membrane (PM) and the endoplasmic reticulum (ER) and find that prewetting is potentiated or inhibited by perturbations of membrane composition. Prewetting can also mediate membrane adhesion, with avidity dependent on membrane composition, as demonstrated in cells through the potentiation or inhibition of ER-PM contact sites. The strong correspondence of results in simulation, reconstitution and cells reveals a new role for membrane lipids in regulating the recruitment and assembly of soluble proteins.
    DOI:  https://doi.org/10.1038/s41589-025-02082-0
  10. Front Neurosci. 2025 ;19 1665272
      The endoplasmic reticulum (ER) and mitochondria are essential organelles that interact closely at specialized sites known as ER-mitochondria-associated membranes (MAMs). MAM is enriched with proteins from both the ER and mitochondria. ER stress sensors-inositol-requiring enzyme 1 (IRE1) and protein kinase RNA-like ER kinase (PERK) - are traditionally recognized for their roles in the unfolded protein response (UPR), which mitigates proteotoxic stress. However, recent studies reveal their non-canonical functions at MAMs, where they regulate calcium signaling, mitochondrial dynamics, and apoptosis through interactions with MAM-resident proteins. Disruption of these pathways is implicated in various diseases, particularly neurodegenerative disorders. This review highlights the emerging roles of IRE1 and PERK in preserving mitochondrial function and their relevance to neurodegeneration. It also examines pharmacological strategies targeting these proteins, which influence both UPR signaling and ER-mitochondrial communication, offering a comprehensive perspective on their roles in health and disease.
    Keywords:  ER stress sensors; ER-mitochondrial interactions; IRE1; UPR signaling; mitochondrial health; neurodegenerative diseases; pERK
    DOI:  https://doi.org/10.3389/fnins.2025.1665272
  11. Front Cell Neurosci. 2025 ;19 1697060
      Traumatic brain injury (TBI) remains a major global public health concern, characterized by high morbidity, mortality, and long-term disability. Beyond the primary mechanical insult, the progression of secondary injuries-including neuroinflammation, oxidative stress, mitochondrial dysfunction, and excitotoxicity-plays a decisive role in long-term neurological outcomes. Emerging evidence positions cellular stress responses at the core of TBI pathophysiology, mediating the transition from acute injury to chronic neurodegeneration. This review systematically outlines the major stress phenotypes triggered by TBI, including oxidative stress, endoplasmic reticulum (ER) stress, mitochondrial distress, and autophagy imbalance. Particular emphasis is placed on the molecular interplay between the mitochondria and ER, where the mitochondria-associated membranes (MAMs) serve as dynamic hubs regulating calcium (Ca2+) homeostasis, ATP production, and apoptotic signaling. Disruptions in Ca2+ flux through MAMs exacerbate energy failure and promote reactive oxygen species (ROS) overproduction, triggering pro-inflammatory cascades and neuronal apoptosis. Furthermore, the crosstalk between ER-mitochondrial stress integrates signals that govern autophagy and inflammatory responses via key nodes such as C/EBP Homologous Protein (CHOP), Nuclear factor erythroid 2-related factor 2(Nrf2), and Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB). We also explore how stress crosstalk mechanistically contributes to neurological dysfunctions, including glial activation, axonal injury, and progressive cognitive-behavioral impairments. Understanding these intricate molecular mechanisms not only elucidates the pathogenesis of secondary brain damage but also unveils novel therapeutic targets for intervention. Targeting stress response integration may represent a transformative approach in preventing long-term disability and enhancing neuroregenerative outcomes following TBI.
    Keywords:  MAMS; TBI; apoptosis and autophagy; calcium signaling; neuroinflammation; oxidative and ER stress
    DOI:  https://doi.org/10.3389/fncel.2025.1697060
  12. Pharmaceuticals (Basel). 2025 Dec 17. pii: 1900. [Epub ahead of print]18(12):
      Pridopidine is a highly selective sigma-1 receptor (S1R) agonist in clinical development for Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS). The S1R is a ubiquitous chaperone protein enriched in the central nervous system and regulates multiple pathways critical for neuronal cell function and survival, including cellular stress responses, mitochondrial function, calcium signaling, protein folding, and autophagy. S1R has a crucial role in the ER mitochondria-associated membrane (MAM), whose dysfunction is implicated in several neurodegenerative diseases. By activating the S1R, pridopidine corrects multiple cellular pathways necessary to the cell's ability to respond to stress, which are disrupted in neurodegenerative diseases. Pridopidine restores MAM integrity; rescues Ca2+ homeostasis and autophagy; mitigates ER stress, mitochondrial dysfunction, and oxidative damage; and enhances brain-derived neurotrophic factor (BDNF) axonal transport and secretion, synaptic plasticity, and dendritic spine density. Pridopidine demonstrates neuroprotective effects in in vivo models of neurodegenerative diseases (NDDs). Importantly, pridopidine demonstrates the biphasic dose response characteristic of S1R agonists. In clinical trials in HD and ALS, pridopidine has shown benefits across multiple endpoints. Pridopidine's mechanism of action, modulating core cellular survival pathways, positions it as a promising candidate for disease modification for different nervous system disorders. Its broad therapeutic potential includes neurodevelopmental disorders, and rare diseases including Wolfram syndrome, Rett syndrome, and Vanishing White Matter Disease. Here, we review the experimental data demonstrating pridopidine's S1R-mediated neuroprotective effects. These findings underscore the therapeutic relevance of S1R activation and support further investigation of pridopidine for the treatment of different neurodegenerative diseases including ALS and HD.
    Keywords:  S1R agonist; Sigma-1 receptor; neurodegenerative disease; neuroprotection; pridopidine
    DOI:  https://doi.org/10.3390/ph18121900
  13. JCI Insight. 2026 Jan 09. pii: e192970. [Epub ahead of print]11(1):
      The activation of the NLRP3 inflammasome is a pivotal step in hyperinflammation in sepsis; however, the regulatory mechanisms underlying its activation are not fully understood. In this study, we found that 14-3-3ε facilitates NLRP3 inflammasome activation by enhancing NLRP3 K63 deubiquitination and promoting its translocation to the mitochondria-associated ER membranes (MAMs) for full activation. Mass spectrometry revealed that 14-3-3ε binds to NLRP3 in macrophages during sepsis. Plasma 14-3-3ε levels were elevated in patients with sepsis and were positively associated with disease severity. 14-3-3ε promoted NLRP3 inflammasome activation by facilitating NLRP3 aggregation and NLRP3-ASC assembly. The interaction between 14-3-3ε and NLRP3 was dependent on phosphorylation at the S194 site of NLRP3 NACHT domain. The NLRP3-14-3-3ε interaction promoted K63 deubiquitination and enhanced the translocation of NLRP3 to MAMs, which is necessary for full activation of NLRP3 inflammasome. Furthermore, macrophage-conditional KO of 14-3-3ε or treatment with BV02, a 14-3-3 inhibitor, improved the survival rate and alleviated organ injuries in septic mice. Taken together, our data indicate that 14-3-3ε functions as a positive regulator of the NLRP3 inflammasome and could be a target for sepsis treatment.
    Keywords:  Infectious disease; Inflammation; Macrophages
    DOI:  https://doi.org/10.1172/jci.insight.192970