JOURNAL OF CELLULAR PHYSIOLOGY

Parathyroid Hormone (1-34) Signaling for Bone Health in Breast Cancer Survivorship and Metastasis
Oh S, Oh HR, Shen C, Park YR and Park JH
Osteoporosis, fragility fractures, and pathologic fractures are increasingly recognized as long-term complications in cancer survivors. Women are more susceptible to bone loss than men, and breast cancer is the most common malignancy in women. In this population, bone health is a critical concern due to both therapy-induced bone loss and a high propensity for skeletal metastasis. Antiresorptive agents are widely used; however, their known adverse effects and limited capacity to rapidly reduce fracture risk in high-risk individuals have led to growing support for the early use of osteoanabolic therapies. Among these, intermittent administration of parathyroid hormone [PTH (1-34)] has demonstrated clinical efficacy in reducing fracture risk by activating the PTH 1 receptor in osteoblasts. However, its safety and mechanistic relevance in the context of breast cancer remain poorly understood. This review outlines the osteoblast-specific signaling pathways of PTH (1-34) and includes our recent research that identified p21-activated kinase 4 as a downstream effector linking canonical cyclic adenosine monophosphate-protein kinase A signaling to Wnt/β-catenin activation. Additionally, it explores the potential implications of PTH (1-34) in the context of breast cancer-related bone metastasis.
A Novel Role of Neutrophil Elastase in Podocyte Dysfunction Induced by High Glucose, PMA, and MDP
Kulesza T, Wróblewska A, Sawicka J, Audzeyenka I, Rachubik P, Rogacka D, Mallek W, Wysocka M, Lesner A and Piwkowska A
Podocytes are not only the key regulators of glomerular filtration barrier dynamics but also exhibit immunological properties. They are capable of antigen presentation and possess a receptor system recognizing pathogen-associated molecular patterns. Our earlier study further demonstrated that podocytes share additional similarities with immune cells, as they can synthesize and secrete the active form of cathepsin C - an enzyme that controls the activation of neutrophil serine proteases (NSPs). In this study, we established that podocytes synthesize neutrophil elastase (NE), proteinase 3 (PR 3), and cathepsin G (CatG) but also release their active forms into the extracellular environment. We found that NSPs contribute to podocyte dysfunction upon inflammation induction by PMA and under conditions of insulin insensitivity. Moreover, actin cytoskeleton rearrangement and increased albumin permeability of the podocyte monolayer were triggered by nucleotide-binding and oligomerization domain-containing protein 2 (NOD2) activation via muramyl dipeptide (MDP), which consequently enhanced NE and PR 3 activity. Notably, silencing the ELANE gene, which encodes NE, exerted a protective effect on podocytes even after NOD2 activation. These findings indicate that NSPs, especially NE, contribute to podocyte dysfunction in diabetes and diabetic kidney disease, a condition characterized by chronic inflammation and insulin resistance.
Telomere Attrition-Induced Senescence in Human Pluripotent Stem Cell-Derived Astrocytes: Distinct Cellular and Functional Characteristics
Kim D, Yoo SH, Oh SS, Yeon GB, Choi J, Lee J, Park S, Kim DW and Kim DS
This study explored the role of telomere attrition in astrocytic senescence by pharmacologically inhibiting telomerase activity in human induced pluripotent stem cell-derived astrocytes. Treatment with the telomerase inhibitor BIBR1532 (BIBR) during differentiation induced hallmark features of senescence, including nuclear lamina abnormalities, enhanced senescence-associated β-galactosidase activity, increased replication arrest and DNA damage, altered reactive oxygen species homeostasis in mitochondria, accompanied by significant shortening of relative telomere length. Despite these senescence related characteristics, BIBR-treated astrocytes exhibited limited changes in the expression of senescence-associated secretory phenotype-related genes. Moreover, their key functional properties, such as glutamate uptake, synaptic vesicle clearance, mitochondrial membrane potential and morphology remain comparable to those of control astrocytes. These findings suggest that the presence of classical senescence markers does not necessarily lead to functional impairment and that BIBR-induced senescence in astrocytes may represent an early or transitional phase, where classical senescence markers emerge without substantial functional decline. Our results reinforce the notion that while telomere attrition is a major cellular senescence driver, its onset may not be attributed to a single stressor but rather to a complex interplay of cellular stress pathways. This study provides valuable insights into the mechanisms underlying astrocytic senescence and underscores the need for further research on the molecular basis of its occurrence and functional implications.
Lactobacillus acidophilus Ameliorates Inflammatory Bone Loss Under Postmenopausal Osteoporotic Conditions via Modulating the Gut-Resident RORγT pTreg Cell Population
Bhardwaj A, Sapra L, Saini C, Tiwari A, Nilakhe A, Mishra PK and Srivastava RK
Research in the past decade has elucidated the explicit role of the immune system in the pathophysiology of osteoporosis. Recent studies have further unraveled the complex interactions between bone and immune cells and explored safe, effective immunomodulatory approaches-such as probiotics-for preventing and managing osteoporosis. As a result, various immune factors have continuously been discovered to play specific roles in maintaining bone homeostasis. The role of Tregs in the context of postmenopausal osteoporosis (PMO) is already well established. While Foxp3 Tregs are mostly matured in the thymus (tTregs), some are also produced from Foxp3CD4 T-cell precursors in the peripheral tissues (i.e., pTregs). Notably, the specific role of pTregs and tTregs in PMO remains to be elucidated. Here, we reveal that estrogen-deficient inflammatory conditions in PMO disrupt the balance of tTregs and pTregs. Interestingly, within pTregs, the population of RORγT pTregs and RORγT pTregs is further altered, along with simultaneous expansion of Th17 cells-likely through the conversion of RORγT pTregs into Th17 cells. Notably, supplementation with Lactobacillus acidophilus (LA) restores the homeostasis of RORγT pTregs and Th17 cells in a butyrate-mediated manner. Moreover, it was observed that butyrate-primed RORγT pTregs have reduced osteoclastogenic potential. Collectively, our findings for the first time reveal the pivotal role of gut resident RORγT pTregs-Th17 cell axis in the pathophysiology of PMO.
Autophagy Reprogramming in Cancer
Fuller AD, Bordner TH, Staub AJ, Jackson JL, Shanas N, Crespo JM, Nazario-Lugo WA, Rukhsar M, Tufano A, Worrell C, Reichenbach ZW and Whelan KA
During malignancy, metabolic reprogramming is critical for cancer cells to survive and thrive in nutrient- and oxygen-poor conditions. Autophagy is a catabolic process through which intracellular components are degraded to support cells upon exposure to stressful conditions. While autophagy is protective during early cancer initiation, tumor cells may initiate cell-intrinsic and cell-extrinsic autophagy to support their survival in later stages of cancer. As autophagy is present at low levels in most tissues under homeostasis and upregulated in malignancy, there has been great interest in targeting the autophagy pathway for cancer therapy. Here, we discuss the mechanisms through which autophagy and autophagy-related proteins act to limit carcinogenesis. We then review pro-tumor roles for autophagy in tumor cells as well as in components of the tumor microenvironment. Finally, we discuss autophagy-targeted approaches for cancer therapy. This review article highlights autophagy as a key player in cell metabolism that is often leveraged to support cancer progression and as a potential therapeutic target in a variety of cancer types.
Cannabinoid Receptors Modulate Physiological Remodelling of the Blood-Testis Barrier
Manfrevola F, Ricci G, Suglia A, Mele VG, Migliaccio A, Chianese R, Cobellis G and Chioccarelli T
The endocannabinoid system, including the CB1 and CB2 receptors, has been associated with the modulation of blood-brain barrier and gut barrier. Herein, using CB1 knock-out male mice, we studied the potential role of these receptors in maintenance of blood-testis barrier (BTB) integrity during the seminiferous epithelium remodelling phase (Stages VIII-XI), focusing on events responsive to CB1 and CB2 activity. Our results showed that the genetic loss of CB1 disrupted testicular expression of some components of BTB, including factors of junctional complexes, promoting tubular infiltration of blood cells. Such infiltration specifically occurred at Stages VIII-IX transition. Gene expression analysis of molecular tags that highlight BTB remodelling (by addressing Occludin to early/late endosome, membrane recycling and proteasome) revealed higher BTB dynamism and impoverishment of tight junctions at Sertoli-Sertoli interface with significant effects on BTB remodelling activities. In detail, CB1 deletion increased kinetic of internalization and recycling of tight junctions and simultaneously promoted proteosome-mediated Occludin degradation with negative effects on permeability of BTB during its remodelling. This caused the leakage of the tight junctions, the premature passage of germ cells in adluminal compartment and downstream the slowing of spermatogenesis. These results strongly indicated that CB1 and CB2 activation contribute to BTB remodelling being both involved in the modulation of tight junction-associated proteins and in their dynamism: these data highlight a new role for CB1 in spermatogenesis.
Mitochondrial and Glucose Metabolic Patterns in Pre-Granulosa Cells and Oocytes and Their Dysfunctions Induce Impaired Primordial Follicle Formation in Mice
Yan Y, Ma W, Li Y, Li J, Zhao X, Wei Q, Peng S, Pan M and Ma B
In mammals, the establishment of primordial follicles (PFs) occurs in an orderly manner and is an energy-demanding process. However, the mechanisms underlying the supply and demand of energy metabolism during primordial follicle formation, particularly glycolysis and oxidative phosphorylation (OXPHOS) signaling, remain poorly understand. Herein, based on the analyses of single-cell RNA sequencing (scRNA-seq) data from mouse ovarian tissues, gene expression associated with glycolysis and OXPHOS signaling were dynamically changed along pseudotime trajectory in pre-granulosa (PG) cells and oocytes following cell development and PF formation. The molecules related to glycolysis and OXPHOS signaling exhibited dynamic expression patterns in mouse ovarian tissues following PF formation, with distinct expression levels and location in somatic cells and oocytes. The dysfunctions of mitochondrial and glucose metabolic patterns using glycolysis inhibitor (2-Deoxy-Dglucose, 2-DG) or OXPHOS signaling inhibitor (metformin, MET) significantly inhibited PF formation, disordered oocyte development, downregulated key gene expression, impaired the recruitment and maintenance of PG cells, and altered cell proliferation and apoptosis. Collectively, these results demonstrate that cellular metabolic patterns are diverse and dynamically regulate in oocytes and PG cells during PF formation of mice, and glucose metabolism is essential for PF formation and its disruption inhibits PF formation.
ELF4/TRIB3/CDK6 Axis Promotes Cancer Stem Cell Activity in Endometrial Cancer
Chen CY, Lee YC, Huang YH, Wang WL and Chang WW
Endometrial cancer (EC) is the most prevalent gynecological malignancy globally. Here, we explored the role of E74-like ETS transcription factor 4 (ELF4) in EC progression. Using the TISIDB web tool to analyze TCGA data, we found that elevated ELF4 expression correlates with higher histological grades and reduced overall survival in EC patients. Tissue microarray analysis confirmed a grade-dependent increase in ELF4 protein levels. Knockdown of ELF4 in EC cell lines (AN3CA, HEC-1A) and patient-derived EC cells suppressed proliferation, cell cycle progression, and cancer stem cell (CSC) activity. Database analysis and RNA interference identified cyclin-dependent kinase 6 (CDK6) as a downstream target of ELF4. ELF4 silencing reduced CDK6 mRNA and protein expression, while chromatin immunoprecipitation revealed direct binding of ELF4 to the CDK6 promoter. Conversely, ELF4 overexpression upregulated CDK6. Knockdown of CDK6 or treatment with the CDK4/6 inhibitor Palbociclib diminished tumorsphere formation and expression of stemness markers (OCT4, NANOG, c-MYC) in both conventional and patient-derived EC cells. We previously reported that the tribbles pseudokinase 3 (TRIB3)/ELF4 complex transactivates CTNNB1 expression; here, we show that TRIB3 knockdown also downregulates CDK6 at mRNA and protein levels, suggesting cooperative regulation of CDK6 by ELF4 and TRIB3. In EC specimens, ELF4, TRIB3, and CDK6 expression positively correlated, and Kaplan-Meier analysis indicated that high co-expression of these genes predicted the poorest overall survival. Collectively, our findings establish the ELF4/TRIB3/CDK6 axis as a critical regulator of EC progression and CSC maintenance, highlighting its potential as a therapeutic target for EC.
From Single-Cell Atlas to Functional Validation: Critical Next Steps for Understanding Tip Cell-Mediated Communication in the Injured Spinal Cord
Yang D, Yang J and Wang G
In their recent study, Zeng et al. (2025) employed single-cell RNA sequencing to delineate the landscape of spinal cord injury (SCI), highlighting a previously underappreciated communicative role for endothelial tip cells in engaging astrocytes and macrophages. While their work provides a valuable resource and generates compelling hypotheses, it also opens several critical avenues that demand immediate scrutiny. This Letter offers a prospective outlook and a critical examination of their findings. We argue that the computationally predicted paracrine networks, such as the Angptl4-Sdc4 axis identified by the authors, require rigorous in vivo functional validation to establish causality. Furthermore, the current snapshot data lack the temporal and spatial resolution necessary to decipher the dynamics of these interactions. Most importantly, we explore the therapeutic dilemma of targeting tip cells-a strategy that must delicately balance their detrimental signaling roles against their indispensable function in revascularization. Addressing these challenges is paramount to transforming these descriptive insights into mechanistic understanding and viable therapeutic strategies for SCI. PRE-REGISTERED CLINICAL TRIAL NUMBER: Not applicable.
Hallmarks of the Aging Skin Microenvironment: Components and Mechanisms
He R, Sun M, Liu T, Geng L, Xing C, Xu P, Peng Y and Fang Y
Skin aging is a complex biological process driven by the dynamic interplay of cellular senescence, molecular dysfunction, and microenvironmental remodeling. The aging microenvironment acts as both a consequence and a driver of skin aging, creating a vicious cycle that exacerbates inflammation, oxidative stress, and barrier dysfunction. An in-depth exploration of the aging skin microenvironment plays a revolutionary role in the field of skin anti-aging and holds promise for the discovery of novel and feasible targets for skin anti-aging. This review systematically elaborates the aging skin microenvironment through a framework of six interconnected components: (1) inflammaging and immune cell dysfunction, (2) extracellular matrix dysregulation, (3) intercellular communication and extracellular vesicles defect, (4) physical microenvironmental alterations, (5) stem cell exhaustion, and (6) microbiome dysbiosis. These components collectively establish a self-reinforcing network that perpetuates structural degradation, functional decline, and impaired regenerative capacity.
Role of Seminal Exosomes in Reproduction
Mehdinejadiani S, Azad N, Dehghan Z, Khosravizadeh Z, Saberi F, Rezaee D, Pilehchi T, Goudarzi N, Salahi E and Mehdinejadiani K
Exosomes are small lipid bilayer vesicles, ranging from 30 to 150 nm in diameter, that are secreted by various cells and facilitate intercellular communication. They originate from the endosomal system and release their contents into the extracellular environment. These nanovesicles carry bioactive molecules, including nucleic acids, lipids, and predominantly proteins, influencing target cells and contributing to cell-to-cell interactions. Exosomes play a crucial role in both normal physiological functions and pathological conditions, including male and female reproductive disorders. Various parts of the male reproductive tract release exosomes into seminal fluid. Seminal exosomes, especially epididymosomes and prostasomes, have been shown to influence male fertility. Furthermore, the role of seminal exosomes has been demonstrated in the female reproductive tract during implantation and pregnancy. Evidence shows that the exosomal cargo in seminal fluid differs between normal and pathological conditions, impacting the reproductive process. Consequently, exosomes are considered valuable biomarkers not only for diagnosis but also for potential therapeutic roles in abnormal conditions, particularly infertility. This review aims to explore the role of seminal exosomes in male fertility and their subsequent impact on the female reproductive tract during fertilization, preimplantation, implantation, postimplantation, and pregnancy-associated diseases, as well as the role of exosomes during seminal infections. Additionally, it aims to highlight the significance of seminal exosomes in medical applications and emphasize the need for future studies in this area.
The Effect and Mechanism of DNAJA1-Mediated Met/Leu Promoting the Proliferation of Bovine Mammary Epithelial Cells
Li Z, Xie Y, Wang Y, Yuan X and Ao J
Milk is a natural product synthesized and secreted by Bovine mammary epithelial cells (BMECs), providing the nutrients needed for the growth and development of calves. At the same time, it is also one of the common beverages in our daily life. The research on the expansion mechanisms of BMECs is of great significance for increasing dairy yield. DNAJA1 pertains to the HSP40 family (alternatively called DNAJ proteins). As an essential mammalian molecular chaperone, this protein features a structurally distinctive J-domain region enabling functional coordination with HSP70 (HSPA). However, the expression mechanism and biological function of DNAJA1 in BMECs remain unclear. This investigation demonstrates DNAJA1's critical involvement in methionine (Met) and leucine (Leu)-modulated BMEC proliferation. Experimental findings reveal that both Met and Leu stimulate BMEC proliferation, with DNAJA1 similarly exerting a positive regulatory influence on cellular multiplication. Within BMECs, Met and Leu augment proliferation by activating the PI3K-AKT-DNAJA1 signaling axis. Concurrently, an interaction between DNAJA1 and TAK1 potentially contributes further to regulating this proliferative process.
Hypoxia Increases Sarcolemmal Na/HCO Cotransport Activity via an ERK1/2-Dependent Pathway in Cardiac HL-1 Cell Line
Şimşek G, Swietach P and Kandilci HB
As a major modulator of cardiac function, intracellular pH (pH) is tightly controlled by sarcolemmal acid-base transporters to within narrow limits (7.1-7.3). Na/H exchanger (NHE1) and Na/HCO cotransporter (NBC) are the main acid-extruding membrane proteins; the latter is further subdivided into electrogenic (NBCe1/NBCe2) and electroneutral (NBCn1) isoforms. In the underperfused heart, acid disturbances are often accompanied by hypoxia, but their interplay on cardiac NBC activity is unknown. Here, we studied the effect of acute (1 mM dithionite and 100% N, 10 min) and long-term hypoxia (1% O, 48 h) on sarcolemmal NBC activity using fluorimetric assays in mouse atrial-derived HL-1 cells and primary rat cardiomyocytes. NBCe1 and NBCn1 transcripts were detected in HL-1 cells. Ensemble NBC activity, defined as the HCO -dependent acid-extrusion flux, was promptly inhibited under acute anoxia. In contrast, pH-sensitivity of NBC flux was increased after long-term hypoxia, likely an adaptive response. This increase was not due to buffering capacity changes but was mimicked by dimethyloxalylglycine (1 mM, DMOG), which stabilizes hypoxia inducible factor under normoxic conditions. Hypoxia affected neither NBCn1 nor NBCe1 protein levels, indicating a modulatory effect on transporter activity. The contribution of electrogenic (NBCe1) and electroneutral (NBCn1) isoforms, dissected from fluxes generated under hyperkalemia, showed that long-term hypoxia selectively raised NBCn1 activity. This effect was blocked by U0126, an inhibitor of extracellular signal-regulated kinase 1/2, implicating phosphorylation. Our results show that acute anoxia and prolonged hypoxia regulate NBC-dependent flux by distinct mechanisms ostensibly to retain pH control under the combination of acidosis and hypoxia.
Endothelial Effects in the Elderly: Fibroblast Regulation in Soft Tissue Healing
Oberringer M, Jennewein M, Bubel M, Guthörl S and Pohlemann T
Two main influencing factors of human soft tissue healing are concomitant diseases and cellular senescence, both accumulating with increasing age. Due to the raising population of the elderly in western countries, it is essential to enhance the level of knowledge concerning the function of senescence in a granulation tissue during repair. The present study was intended to verify classic markers of senescence, like senescence-associated ß-galactosidase and the development of a senescence-associated secretory phenotype among fibroblasts during emerging senescence. The application of an in vitro model using serial passaging as inducer of replicative senescence revealed specific differences of a non-senescent and a pre-senescent fibroblast phenotype in mono-cultures, representing the basis for a detailed examination of the phenotypes in their interaction with microvascular endothelial cells in co-cultures. The results deliver new insights into the age dependent process of tissue repair. Characteristics of pre-senescent fibroblasts in terms of modified proliferation, cell morphology, cell cycle regulation, myofibroblastoid differentiation and cytokine release indicate a strong responsibility of this phenotype for the composition and function of a granulation tissue at different locations, including vascular sites. In its entirety, the results support the assumption, that a missing clearance of the senescence phenotype in late stages of tissue repair is one of the main reasons for healing failure.
The Role of GPR39 in Regulating Osteoblast Function, Bone Matrix Quality, and Gender-Specific Bone Homeostasis
Chaterjee B, Gozlan G, Abramovitch-Dahan C, Davydok A, Reiner-Benaim A, Krug J, Jähn-Rickert K, Busse B and Levaot N
GPR39, a zinc-sensing receptor, is essential for bone homeostasis in male mice through regulation of osteoblast function and matrix composition. This study examined the effects of GPR39 deficiency in female mice using both global and osteoblast lineage-specific GPR39 knockout models (Gpr39). In vivo, GPR39-deficient female mice exhibited reduced bone mass, increased mineralization rates, and significantly lower and more variable serum levels of pro-collagen type I N-propeptide (PINP), indicating impaired collagen synthesis and matrix remodeling. OVX models further demonstrated that GPR39 deficiency exacerbates estrogen-deficiency-induced bone loss, highlighting its protective role in postmenopausal-like states. Osteoblast lineage-specific GPR39 deletion replicated the skeletal abnormalities observed in global knockouts, revealing that GPR39 activity in the osteoblast lineage is indispensable for proper collagen deposition and mineralization. Western blot analysis of Gpr39 osteoblasts confirmed reduced extracellular collagen levels, while quantitative mRNA analysis of Col1a2 revealed zinc signaling through GPR39 as a key regulator of collagen production. Zinc-induced Col1a2 expression, dependent on GPR39 and mediated via Gα signaling, was abolished in GPR39-deficient osteoblasts. These findings provide insights into how zinc signaling via GPR39 regulates osteoblast function and collagen synthesis, emphasizing its role in maintaining matrix composition. Targeting GPR39 may offer novel therapeutic strategies for osteoporosis and other bone disorders characterized by impaired matrix remodeling.
CD200 Promotes Gastric Cancer Progression and Metastasis by Inducing the β-catenin Signaling Pathway
Lee H, Bae JM, Shin SP, Kim W, Kim WJ, Kang HG, Choi DB, Lee YS and Kim SJ
Gastric cancer is the fifth most common malignancy and the fourth leading cause of cancer-related mortalities worldwide. Understanding the mechanisms driving tumor growth and metastasis in gastric cancer is essential for the development of effective therapeutic strategies. In this regard, it is well-established that CD200, a glycoprotein that binds to the CD200 receptor, has notable immunosuppressive effects. The extracellular domain of CD200 is secreted into the tumor microenvironment (TME), wherein it promotes cancer progression. However, although CD200 is highly expressed in several types of cancers, the details of its intracellular roles in tumor progression remain poorly understood. In this study, we investigated the biological function and mechanism of action of CD200 in gastric cancer. Public datasets from GSE and TCGA revealed that CD200 is overexpressed in gastric cancer and that its expression is correlated with cancer stage and metastasis. Functionally, we found that CD200 enhances cell proliferation, migration, and invasion, and also promotes the expression of epithelial-mesenchymal transition (EMT)-related genes. Mechanistically, CD200 was demonstrated activate the WNT/β-catenin signaling pathway by inducing β-catenin activation. Notably, we established that the cytoplasmic domain of CD200 binds directly to β-catenin, thereby facilitating its nuclear translocation. The CD200/β-catenin/TCF4 complex subsequently promotes the transcription of β-catenin target and EMT-related genes. Collectively, our findings in this study revealed that the cytoplasmic domain of CD200 interacts with β-catenin, thereby promoting the transcriptional activation of β-catenin target genes and inducing tumor growth and metastasis in gastric cancer. These findings accordingly indicate that CD200 may serve as a potential therapeutic target for the treatment of gastric cancer.
Bidirectional Regulatory Mechanisms of Disulfidptosis Govern Precision Targeting in Degenerative Bone Diseases and Osteosarcoma
Zhao X, Wu S, Su X, Gao J, Qu L, Zhang Y and Zhang Y
Disulfidptosis is a newly identified form of programmed cell death closely associated with cystine metabolism abnormalities and cytoskeletal damage. Orthopedic diseases, such as degenerative conditions including intervertebral disc degeneration, osteoporosis, osteoarthritis, and malignant bone tumors like osteosarcoma, all involve imbalances in the immunometabolic microenvironment. The triggering conditions for disulfidptosis, such as high expression of SLC7A11 and glucose deprivation, are highly correlated with the pathaological features of orthopedic diseases and associated immune dysregulation. However, there is currently a lack of systematic understanding regarding the regulatory networks, molecular markers, and intervention strategies of disulfidptosis in orthopedic diseases, and the specific mechanisms by which it contributes to disease onset and progression remain unclear. This review systematically summarizes the bidirectional immunometabolic regulatory molecular mechanisms, pathological associations, and potential therapeutic strategies of disulfidptosis in orthopedic degenerative diseases and bone tumors. By analyzing the immunometabolic regulatory networks of key molecules such as SLC7A11, TXNRD1, and RPN1, we propose immune-aware precision strategies combining disulfidptosis-targeted metabolic intervention with checkpoint blockade immunotherapy. This review fills the gap in the research of disulfidptosis in orthopedic diseases, providing new insights for a deeper understanding of the molecular mechanisms underlying these conditions, while establishing a theoretical framework for developing precise therapeutic strategies based on the regulation of disulfidptosis.
Mettl14 Deficiency Promotes Fam32a Expression via m6A Modifications to Facilitate the Hepatocyte G1/S Transition
Li C, Zhou W, Wang D, Liu Z, Shen M, Zhu L, Li H, Zhong X and Liu H
Mettl14, a key component of the m6A methyltransferase complex, plays a crucial role in regulating mRNA stability and splicing. Reduced expression of Mettl14 is associated with hepatocellular carcinoma and liver regeneration, yet the molecular mechanisms by which it regulates the hepatocyte cell cycle remain unclear. Using RNA-Seq and MeRIP-Seq in liver-specific Mettl14 knockout mice, we found that Mettl14 deficiency stabilizes Fam32a mRNA through m6A modifications, resulting in increased Fam32a protein levels. Elevated Fam32a expression accelerates the G1/S transition by modulating Cdkn1a splicing, specifically downregulating its variant 2. These findings uncover a novel m6A-dependent mechanism that regulates hepatocyte cell cycle progression and highlight the previously unrecognized role of Fam32a in promoting the G1/S transition.
Multidrug Resistance-Associated Protein 1 Aberration-Incurred Glutathione Efflux Drives Renal Ferroptosis and Acute Kidney Injury-Chronic Kidney Disease Progression
Zhao J, An H, Zhou S, Tian T, Yao Y and Shi L
The progression of acute kidney injury (AKI) to chronic kidney disease (CKD) represents a unique renal disease scenario, yet its exact mechanisms remain unclear. The transport of renal metabolic byproducts plays a crucial role in maintaining systemic homeostasis and the repair process. The glutathione-based lipid oxidation-reduction system is essential for preserving cellular function. However, the relationship between the disruption of the redox system during the AKI-CKD transition and renal transport proteins remains unclear. We investigated the mechanisms by which the transport protein multidrug resistance-associated protein 1 (MRP1) mediates the destruction of the redox system during renal ischemia-reperfusion injury (IRI) and devised interventions related to renal ferroptosis. Transcriptome analysis and a unilateral kidney IRI model were employed to explore changes in MRP1 expression during the AKI-CKD process. Functional experiments simulating in vivo renal IRI were conducted using Carbonyl Cyanide m-Chlorophenylhydrazine (CCCP)-treated renal tubular epithelial cells. MK571(MRP1 inhibitor) and Fer-1 were used to inhibit MRP1 and ferroptosis, respectively. Kidney tissue damage and fibrosis area were evaluated using staining methods like KIM1 and Masson. In the renal IRI model, upregulation of the transport protein MRP1 expression in renal tissue was observed. MRP1 is responsible for transporting glutathione outside the cell. MK571 significantly inhibited the AKI- CKD transition and immune cell infiltration. Both the deletion or inhibition of MRP1 can also alleviate ferroptosis. However, the combined use of MK571 and Fer-1 did not show additional kidney protective effects. Elevated expression of the renal transport protein MRP1 during renal IRI induces the extracellular leakage of glutathione, leading to ferroptosis. Inhibiting MRP1 can slow down renal ferroptosis and the progression from AKI-CKD.
Overeating: A Conundrum of Lipid Metabolism and Inflammation in Obesity and Cancer
Surumbayeva A, Saiman Y and Astsaturov I
The global rise in obesity has coincided with an alarming increase in early-onset cancers and other chronic noncommunicable diseases, underscoring the urgent need to understand the underlying mechanisms linking excess body weight with disease pathogenesis. While genetic factors account for disease risk, environmental and dietary influences, particularly those associated with Western hypercaloric diets, play a dominant role in shaping metabolic health. Obesity-driven insulin resistance and chronic inflammation are now recognized as central contributors to a wide range of pathologies, including cardiovascular disease, type 2 diabetes, fatty liver, and multiple cancer types. Emerging evidence suggests that disrupted insulin signaling, altered lipid metabolism, and chronic inflammation converge to promote a tumor-permissive tissue microenvironment. This review examines the mechanistic links between insulin signaling, lipid metabolism, and inflammation as the causality of increased cancer risk in obesity.
Holding Out for a Model: Rhomboid Superfamily in Vertebrate Development and Disease
Gourkanti S, Munoz Y, Cheung J, Chavez RM, Agarwal D, Schoen TJ, Solorio-Kirpichyan K and Neal SE
The rhomboid superfamily, comprising both proteases and pseudoproteases, has emerged as a central regulator of membrane biology, mediating diverse functions including protein quality control, signal transduction, trafficking, and more. While molecular mechanisms of rhomboid activity have been well-characterized in invertebrate and cell-based systems, their physiological role in vertebrate development remains limited and continues to evolve. Here, we review recent advances in cell culture systems and vertebrate models that uncover the developmental and disease-relevant functions of rhomboid family members, including RHBDLs, iRhoms, PARL, and Derlins. We outline their roles in embryogenesis, tissue regeneration, neurodevelopment, and immune signaling, alongside their pathological involvement in cancer, neurodegeneration, and metabolic disorders. We also emphasize the limitations posed by early embryonic lethality in knockout models and advocate for tissue-specific vertebrate models to dissect rhomboid-dependent pathways in vivo. Understanding how rhomboid proteins coordinate developmental processes will not only reveal fundamental principles of membrane-associated processes, but also open new avenues for therapeutic targeting in disease.