Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis Rep ; 8(1): 903-922, 2024.
Article in English | MEDLINE | ID: mdl-38910936

ABSTRACT

Background: Despite intense investigations, no effective treatment is yet available to reduce plaques and protect memory and learning in patients with Alzheimer's disease (AD), the most common neurodegenerative disorder. Therefore, it is important to identify a non-toxic, but effective, treatment option for AD. Objective: Cinnamein, a nontoxic compound, is naturally available in Balsam of Peru and Tolu Balsam. We examined whether cinnamein treatment could decrease plaques and improve cognitive functions in 5XFAD mouse model of AD. Methods: We employed in silico analysis, time-resolved fluorescence energy transfer assay, thermal shift assay, primary neuron isolation, western blot, immunostaining, immunohistochemistry, Barnes maze, T maze, and open field behavior. Results: Oral administration of cinnamein led to significant reduction in amyloid-ß plaque deposits in the brain and protection of spatial learning and memory in 5XFAD mice. Peroxisome proliferator-activated receptor alpha (PPARα), a nuclear hormone receptor, is involved in plaque lowering and increase in hippocampal plasticity. While investigating underlying mechanisms, we found that cinnamein served as a ligand of PPARα. Accordingly, oral cinnamein upregulated the level of PPARα, but not PPARß, in the hippocampus, and remained unable to decrease plaques from the hippocampus and improve memory and learning in 5XFAD mice lacking PPARα. While A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) is one of the drivers of nonamyloidogenic pathway, transcription factor EB (TFEB) is considered as the master regulator of autophagy. Cinnamein treatment was found to upregulate both ADAM10 and TFEB in the brain of 5XFAD mice via PPARα. Conclusions: Our results suggest that this balsam component may have therapeutic importance in AD.

2.
J Neurochem ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38419348

ABSTRACT

Oligodendrocytes are the myelinating cells in the CNS and multiple sclerosis (MS) is a demyelinating disorder that is characterized by progressive loss of myelin. Although oligodendroglial progenitor cells (OPCs) should be differentiated into oligodendrocytes, for multiple reasons, OPCs fail to differentiate into oligodendrocytes in MS. Therefore, increasing the maturation of OPCs to oligodendrocytes may be of therapeutic benefit for MS. The ß-hydroxy ß-methylbutyrate (HMB) is a muscle-building supplement in humans and this study underlines the importance of HMB in stimulating the maturation of OPCs to oligodendrocytes. HMB treatment upregulated the expression of different maturation markers including PLP, MBP, and MOG in cultured OPCs. Double-label immunofluorescence followed by immunoblot analyses confirmed the upregulation of OPC maturation by HMB. While investigating mechanisms, we found that HMB increased the maturation of OPCs isolated from peroxisome proliferator-activated receptor ß-/- (PPARß-/- ) mice, but not PPARα-/- mice. Similarly, GW6471 (an antagonist of PPARα), but not GSK0660 (an antagonist of PPARß), inhibited HMB-induced maturation of OPCs. GW9662, a specific inhibitor of PPARγ, also could not inhibit HMB-mediated stimulation of OPC maturation. Furthermore, PPARα agonist GW7647, but neither PPARß agonist GW0742 nor PPARγ agonist GW1929, alone increased the maturation of OPCs. Finally, HMB treatment of OPCs led to the recruitment of PPARα, but neither PPARß nor PPARγ, to the PLP gene promoter. These results suggest that HMB stimulates the maturation of OPCs via PPARα and that HMB may have therapeutic prospects in remyelination.

3.
Nutr Neurosci ; 27(3): 271-288, 2024 Mar.
Article in English | MEDLINE | ID: mdl-36947578

ABSTRACT

OBJECTIVES: In this study mice were fed a high-fat diet for 12 weeks to establish diet-induced obesity and syringic acid (SA) was assessed for anti-obese, neuroprotective, and neurogenesis. METHOD: Animals were given HFD for 12 weeks to measure metabolic characteristics and then put through the Barns-maze and T-maze tests to measure memory. Additionally, the physiology of the blood-brain barrier, oxidative stress parameters, the expression of inflammatory genes, neurogenesis, and histopathology was evaluated in the brain. RESULT: DIO raised body weight, BMI, and other metabolic parameters after 12 weeks of overfeeding. A reduced spontaneous alternation in behavior (working memory, reference memory, and total time to complete a task), decreased enzymatic and non-enzymatic antioxidants, oxidative biomarkers, increased neurogenesis, and impaired blood-brain barrier were all seen in DIO mice. SA (50 mg/kg) treatment of DIO mice (4 weeks after 8 weeks of HFD feeding) reduced diet-induced changes in lipid parameters associated with obesity, hepatological parameters, memory, blood-brain barrier, oxidative stress, neuroinflammation, and neurogenesis. SA also reduced the impact of malondialdehyde and enhanced the effects of antioxidants such as glutathione, superoxide dismutase (SOD), and total thiol (MDA). Syringic acid improved neurogenesis, cognition, and the blood-brain barrier while reducing neurodegeneration in the hippocampal area. DISCUSSION: According to the results of the study, syringic acid therapy prevented neurodegeneration, oxidative stress, DIO, and memory loss. Syringic acid administration may be a useful treatment for obesity, memory loss, and neurogenesis, but more research and clinical testing is needed.


Subject(s)
Gallic Acid/analogs & derivatives , Obesity , Overweight , Mice , Animals , Obesity/etiology , Obesity/prevention & control , Obesity/drug therapy , Brain/metabolism , Antioxidants/pharmacology , Diet, High-Fat/adverse effects , Oxidative Stress , Memory Disorders , Neurogenesis/physiology
5.
ACS Appl Bio Mater ; 7(1): 44-58, 2024 01 15.
Article in English | MEDLINE | ID: mdl-38108852

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a fundamental process driving cancer metastasis, transforming non-motile cells into a motile population that migrates to distant organs and forms secondary tumors. In recent years, cancer research has revealed a strong connection between exosomes and the EMT. Exosomes, a subpopulation of extracellular vesicles, facilitate cellular communication and dynamically regulate various aspects of cancer metastasis, including immune cell suppression, extracellular matrix remodeling, metastasis initiation, EMT initiation, and organ-specific metastasis. Tumor-derived exosomes (TEXs) and their molecular cargo, comprising proteins, lipids, nucleic acids, and carbohydrates, are essential components that promote EMT in cancer. TEXs miRNAs play a crucial role in reprogramming the tumor microenvironment, while TEX surface integrins contribute to organ-specific metastasis. Exosome-based cancer metastasis research offers a deeper understanding about cancer and an effective theranostic platform development. Additionally, various therapeutic sources of exosomes are paving the way for innovative cancer treatment development. In this Review, we spotlight the role of exosomes in EMT and their theranostic impact, aiming to inspire cancer researchers worldwide to explore this fascinating field in more innovative ways.


Subject(s)
Exosomes , Extracellular Vesicles , MicroRNAs , Neoplasms , Humans , Exosomes/genetics , Exosomes/metabolism , Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Epithelial-Mesenchymal Transition , Tumor Microenvironment
6.
J Med Virol ; 95(10): e29135, 2023 10.
Article in English | MEDLINE | ID: mdl-37792364

ABSTRACT

Personalized cancer immunotherapies, combined with nanotechnology (nano-vaccines), are revolutionizing cancer treatment strategies, explicitly targeting Human papilloma virus (HPV)-related cancers. Despite the availability of preventive vaccines, HPV-related cancers remain a global concern. Personalized cancer nano-vaccines, tailored to an individual's tumor genetic mutations, offer a unique and promising solution. Nanotechnology plays a critical role in these vaccines by efficiently delivering tumor-specific antigens, enhancing immune responses, and paving the way for precise and targeted therapies. Recent advancements in preclinical models have demonstrated the potential of polymeric nanoparticles and high-density lipoprotein-mimicking nano-discs in augmenting the efficacy of personalized cancer vaccines. However, challenges related to optimizing the nano-carrier system and ensuring safety in human trials persist. Excitingly, the integration of nanotechnology with Proteolysis-Targeting Chimeras (PROTACs) provides an additional avenue to enhance the effectiveness of personalized cancer treatment. PROTACs selectively degrade disease-causing proteins, amplifying the impact of nanotechnology-based therapies. Overcoming these challenges and leveraging the synergistic potential of nanotechnology, PROTACs, and Proteolysis-Targeting Antibodies hold great promise in pursuing novel and effective therapeutic solutions for individuals affected by HPV-related cancers.


Subject(s)
Cancer Vaccines , Neoplasms , Papillomavirus Infections , Humans , Proteolysis Targeting Chimera , Human Papillomavirus Viruses , Papillomavirus Infections/complications , Papillomavirus Infections/drug therapy , Proteolysis , Neoplasms/therapy
7.
ACS Omega ; 8(40): 36614-36627, 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37841156

ABSTRACT

Ovarian cancer (OC) is a common gynecological cancer worldwide. Unfortunately, the lack of early detection methods translates into a substantial cohort of women grappling with the pressing health crisis. The discovery of extracellular vesicles (EVs) (their major subpopulation exosomes, microvesicles, and apoptotic bodies) has provided new insights into the understanding of cancer. Exosomes, a subpopulation of EVs, play a crucial role in cellular communication and reflect the cellular status under both healthy and pathological conditions. Tumor-derived exosomes (TEXs) dynamically influence ovarian cancer progression by regulating uncontrolled cell growth, immune suppression, angiogenesis, metastasis, and the development of drug and therapeutic resistance. In the field of OC diagnostics, TEXs offer potential biomarkers in various body fluids. On the other hand, exosomes have also shown promising abilities to cure ovarian cancer. In this review, we address the interlink between exosomes and ovarian cancer and explore their theragnostic signature. Finally, we highlight future directions of exosome-based ovarian cancer research.

8.
NeuroImmune Pharm Ther ; 2(3): 293-300, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37885917

ABSTRACT

Objectives: Although microglia are activated in adult and aged brains resulting in neurodegenerative and neuroinflammatory disorders, most of the cell culture studies on microglia deal with neonatal microglia because of ease of isolation. Microglia could be isolated from adult brains, but it requires separation by density gradient centrifugation, magnetic beads, etc. Here, we describe a simple protocol of isolating highly purified microglia from adult mouse brains. Methods: Our protocol involves dilution with sterile PBS or media, regular centrifugation, and plating on poly-D-lysine-coated flasks. Results: These adult microglia expressed the inducible nitric oxide synthase in response to preformed α-syn fibril, an etiological reagent of Parkinson's disease, and bacterial lipopolysaccharides, one of the prototype proinflammatory stimuli. Moreover, these adult microglia exhibited phagocytosis, which was stimulated by LPS treatment. Conclusions: These results suggest that adult microglia isolated by our procedure are functional and that these adult microglia could be used for studies related to neurodegenerative disorders.

9.
ACS Chem Neurosci ; 14(17): 2981-2994, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37624044

ABSTRACT

Traumatic brain injury (TBI) is a common cause of disability and fatality worldwide. Depending on the clinical presentation, it is a type of acquired brain damage that can be mild, moderate, or severe. The degree of patient's discomfort, prognosis, therapeutic approach, survival rates, and recurrence can all be strongly impacted by an accurate diagnosis made early on. The Glasgow Coma Scale (GCS), along with neuroimaging (MRI (Magnetic Resonance Imaging) and CT scan), is a neurological assessment tools used to evaluate and categorize the severity of TBI based on the patient's level of consciousness, eye opening, and motor response. Extracellular vesicles (EVs) are a growing domain, explaining neurological complications in a more detailed manner. EVs, in general, play a role in cellular communication. Its molecular signature such as DNA, RNA, protein, etc. contributes to the status (health or pathological stage) of the parental cell. Brain-derived EVs support more specific screening (diagnostic and prognostic) in TBI research. Therapeutic impact of EVs are more promising for aiding in TBI healing. It is nontoxic, biocompatible, and capable of crossing the blood-brain barrier (BBB) to transport therapeutic molecules. This review has highlighted the relationships between EVs and TBI theranostics, EVs and TBI-related clinical trials, and related research domain-associated challenges and solutions. This review motivates further exploration of associations between EVs and TBI and develops a better approach to TBI management.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Extracellular Vesicles , Humans , Brain , Blood-Brain Barrier
10.
ACS Biomater Sci Eng ; 9(9): 5205-5221, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37578350

ABSTRACT

Glioblastoma (GBM) is an aggressive type of cancer that has led to the death of a large population. The traditional approach fails to develop a solution for GBM's suffering life. Extensive research into tumor microenvironments (TME) indicates that TME extracellular vesicles (EVs) play a vital role in cancer development and progression. EVs are classified into microvacuoles, apoptotic bodies, and exosomes. Exosomes are the most highlighted domains in cancer research. GBM cell-derived exosomes participate in multiple cancer progression events such as immune suppression, angiogenesis, premetastatic niche formation (PMN), ECM (extracellular matrix), EMT (epithelial-to-mesenchymal transition), metastasis, cancer stem cell development and therapeutic and drug resistance. GBM exosomes also carry the signature of a glioblastoma-related status. The exosome-based GBM examination is part of the new generation of liquid biopsy. It also solved early diagnostic limitations in GBM. Traditional therapeutic approaches do not cross the blood-brain barrier (BBB). Exosomes are a game changer in GBM treatment and it is emerging as a potential platform for effective, efficient, and specific therapeutic development. In this review, we have explored the exosome-GBM interlink, the clinical impact of exosomes on GBM biomarkers, the therapeutics signature of exosomes in GBM, exosome-based research challenges, and future directions in GBM. Therefore, the GBM-derived exosomes offer unique therapeutic opportunities, which are currently under preclinical and clinical testing.


Subject(s)
Exosomes , Extracellular Vesicles , Glioblastoma , Humans , Glioblastoma/therapy , Glioblastoma/diagnosis , Glioblastoma/pathology , Exosomes/pathology , Precision Medicine , Extracellular Vesicles/pathology , Cell Differentiation , Tumor Microenvironment
11.
ACS Appl Bio Mater ; 6(7): 2576-2590, 2023 07 17.
Article in English | MEDLINE | ID: mdl-37314223

ABSTRACT

Cancer is a complex deadly disease that has caused a global health crisis in recent epochs. Colorectal cancer (CRC) is the third most common malignant gastrointestinal disease. It has led to high mortality due to early diagnostic failure. Extracellular vesicles (EVs) come with promising solutions for CRC. Exosomes (a subpopulation of EVs) play a vital role as signaling molecules in CRC tumor microenvironment. It is secreted from all active cells. Exosome-based molecular transport (DNA, RNA, proteins, lipids, etc.) transforms the recipient cell's nature. In CRC, tumor cell-derived exosomes (TEXs) regulate multiple events of CRC development and progression such as immunogenic suppression, angiogenesis, epithelial-mesenchymal transitions (EMT), physical changes in the extracellular matrix (ECM), and metastasis. Biofluid-circulated tumor-derived exosomes (TEXs) are a potential tool for CRC liquid biopsy. Exosome-based colorectal cancer detection creates a great impact in CRC biomarker research. The exosome-associated CRC theranostics approach is a state-of-the-art method. In this review, we address the CRC and exosomes complex associated with cancer development and progression, the impact of exosomes on CRC screening (diagnostic and prognostic biomarkers), and also highlight several exosomes with CRC clinical trials, as well as future directions of exosome-based CRC research. Hopefully, it will encourage several researchers to develop a potential exosome-based theranostic tool to fight CRC.


Subject(s)
Colorectal Neoplasms , Exosomes , Extracellular Vesicles , Humans , Exosomes/genetics , Exosomes/metabolism , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/metabolism , Signal Transduction , Tumor Microenvironment
12.
J Immunol ; 211(2): 187-198, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37314416

ABSTRACT

Although several immunomodulatory drugs are available for multiple sclerosis (MS), most present significant side effects with long-term use. Therefore, delineation of nontoxic drugs for MS is an important area of research. ß-Hydroxy ß-methylbutyrate (HMB) is accessible in local GNC stores as a muscle-building supplement in humans. This study underlines the importance of HMB in suppressing clinical symptoms of experimental autoimmune encephalomyelitis (EAE) in mice, an animal model of MS. Dose-dependent study shows that oral HMB at a dose of 1 mg/kg body weight/d or higher significantly suppresses clinical symptoms of EAE in mice. Accordingly, orally administered HMB attenuated perivascular cuffing, preserved the integrity of the blood-brain barrier and blood-spinal cord barrier, inhibited inflammation, maintained the expression of myelin genes, and blocked demyelination in the spinal cord of EAE mice. From the immunomodulatory side, HMB protected regulatory T cells and suppressed Th1 and Th17 biasness. Using peroxisome proliferator-activated receptor (PPAR)α-/- and PPARß-/- mice, we observed that HMB required PPARß, but not PPARα, to exhibit immunomodulation and suppress EAE. Interestingly, HMB reduced the production of NO via PPARß to protect regulatory T cells. These results describe a novel anti-autoimmune property of HMB that may be beneficial in the treatment of MS and other autoimmune disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , PPAR-beta , Humans , Mice , Animals , PPAR-beta/therapeutic use , Multiple Sclerosis/drug therapy , Valerates/therapeutic use , Mice, Inbred C57BL
13.
Biomarkers ; 28(6): 502-518, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37352015

ABSTRACT

Breast cancer (BC) remains the most challenging global health crisis of the current decade, impacting a large population of females annually. In the field of cancer research, the discovery of extracellular vesicles (EVs), specifically exosomes (a subpopulation of EVs), has marked a significant milestone. In general, exosomes are released from all active cells but tumour cell-derived exosomes (TDXs) have a great impact (TDXs miRNAs, proteins, lipid molecules) on cancer development and progression. TDXs regulate multiple events in breast cancer such as tumour microenvironment remodelling, immune cell suppression, angiogenesis, metastasis (EMT-epithelial mesenchymal transition, organ-specific metastasis), and therapeutic resistance. In BC, early detection is the most challenging event, exosome-based BC screening solved the problem. Exosome-based BC treatment is a sign of the transforming era of liquid biopsy, it is also a promising therapeutic tool for breast cancer. Exosome research goes to closer precision oncology via a single exosome profiling approach. Our hope is that this review will serve as motivation for researchers to explore the field of exosomes and develop an efficient, and affordable theranostics approach for breast cancer.


Subject(s)
Breast Neoplasms , Exosomes , MicroRNAs , Female , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Precision Medicine , Clinical Relevance , Exosomes/genetics , Exosomes/pathology , Tumor Microenvironment/genetics
15.
Phytother Res ; 37(7): 3097-3120, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37246823

ABSTRACT

Phytoestrogens are plant secondary metabolite that is structurally and functionally similar to mammalian estrogens, which have been shown to have various health benefits in humans. Isoflavones, coumestans, and lignans are the three major bioactive classes of phytoestrogens. It has a complicated mechanism of action involving an interaction with the nuclear estrogen receptor isoforms ERα and ERß, with estrogen agonist and estrogen antagonist effects. Depending on their concentration and bioavailability in various plant sources, phytoestrogens can act as estrogen agonist or antagonists. Menopausal vasomotor symptoms, breast cancer, cardiovascular disease, prostate cancer, menopausal symptoms, and osteoporosis/bone health have all been studied using phytoestrogens as an additional standard hormone supplemental remedy. The botanical sources, techniques of identification, classification, side effects, clinical implications, pharmacological and therapeutic effects of their proposed mode of action, safety issues, and future directions for phytoestrogens have all been highlighted in this review.


Subject(s)
Breast Neoplasms , Isoflavones , Prostatic Neoplasms , Animals , Male , Humans , Phytoestrogens/pharmacology , Phytoestrogens/therapeutic use , Estrogens/therapeutic use , Receptors, Estrogen/metabolism , Breast Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Isoflavones/pharmacology , Isoflavones/therapeutic use , Mammals/metabolism
16.
J Cell Mol Med ; 27(11): 1603-1607, 2023 06.
Article in English | MEDLINE | ID: mdl-37183560

ABSTRACT

This short communication will enlighten the readers about the exosome and the epithelial-mesenchymal transition (EMT) related to several complicated events. It also highlighted the therapeutic potential of exosomes against EMT. Exosome toxicology, exosome heterogeneity, and a single exosome profiling approach are also covered in this article. In the future, exosomes could help us get closer to cancer vaccine and precision oncology.


Subject(s)
Exosomes , Neoplasms , Humans , Epithelial-Mesenchymal Transition , Signal Transduction , Precision Medicine
17.
ACS Biomater Sci Eng ; 9(2): 577-594, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36621949

ABSTRACT

Exosomes are the phospholipid-membrane-bound subpopulation of extracellular vesicles derived from the plasma membrane. The main activity of exosomes is cellular communication. In cancer, exosomes play an important rolefrom two distinct perspectives, one related to carcinogenesis and the other as theragnostic and drug delivery tools. The outer phospholipid membrane of Exosome improves drug targeting efficiency. . Some of the vital features of exosomes such as biocompatibility, low toxicity, and low immunogenicity make it a more exciting drug delivery system. Exosome-based drug delivery is a new innovative approach to cancer treatment. Exosome-associated biomarker analysis heralded a new era of cancer diagnostics in a more specific way. This Review focuses on exosome biogenesis, sources, isolation, interrelationship with cancer and exosome-related cancer biomarkers, drug loading methods, exosome-based biomolecule delivery, advances and limitations of exosome-based drug delivery, and exosome-based drug delivery in clinical settings studies. The exosome-based understanding of cancer will change the diagnostic and therapeutic approach in the future.


Subject(s)
Exosomes , Extracellular Vesicles , Neoplasms , Humans , Exosomes/metabolism , Precision Medicine , Drug Delivery Systems/methods , Extracellular Vesicles/metabolism , Neoplasms/diagnosis , Neoplasms/drug therapy , Phospholipids/metabolism , Phospholipids/therapeutic use
19.
J Biol Chem ; 297(5): 101303, 2021 11.
Article in English | MEDLINE | ID: mdl-34655614

ABSTRACT

As a central player in the canonical TGF-ß signaling pathway, Smad2 transmits the activation of TGF-ß receptors at the plasma membrane (PM) to transcriptional regulation in the nucleus. Although it has been well established that binding of TGF-ß to its receptors leads to the recruitment and activation of Smad2, the spatiotemporal mechanism by which Smad2 is recruited to the activated TGF-ß receptor complex and activated is not fully understood. Here we show that Smad2 selectively and tightly binds phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) in the PM. The PI(4,5)P2-binding site is located in the MH2 domain that is involved in interaction with the TGF-ß receptor I that transduces TGF-ß-receptor binding to downstream signaling proteins. Quantitative optical imaging analyses show that PM recruitment of Smad2 is triggered by its interaction with PI(4,5)P2 that is locally enriched near the activated TGF-ß receptor complex, leading to its binding to the TGF-ß receptor I. The PI(4,5)P2-binding activity of Smad2 is essential for the TGF-ß-stimulated phosphorylation, nuclear transport, and transcriptional activity of Smad2. Structural comparison of all Smad MH2 domains suggests that membrane lipids may also interact with other Smad proteins and regulate their function in diverse TGF-ß-mediated biological processes.


Subject(s)
Phosphatidylinositol 4,5-Diphosphate/metabolism , Signal Transduction , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , Active Transport, Cell Nucleus , HeLa Cells , Humans , Phosphatidylinositol 4,5-Diphosphate/genetics , Protein Binding , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Smad2 Protein/genetics , Transforming Growth Factor beta/genetics
20.
Mol Cell ; 71(6): 1092-1104.e5, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30174291

ABSTRACT

Activation of class I phosphatidylinositol 3-kinase (PI3K) leads to formation of phosphatidylinositol-3,4,5-trisphophate (PIP3) and phosphatidylinositol-3,4-bisphophate (PI34P2), which spatiotemporally coordinate and regulate a myriad of cellular processes. By simultaneous quantitative imaging of PIP3 and PI34P2 in live cells, we here show that they have a distinctively different spatiotemporal distribution and history in response to growth factor stimulation, which allows them to selectively induce the membrane recruitment and activation of Akt isoforms. PI34P2 selectively activates Akt2 at both the plasma membrane and early endosomes, whereas PIP3 selectively stimulates Akt1 and Akt3 exclusively at the plasma membrane. These spatiotemporally distinct activation patterns of Akt isoforms provide a mechanism for their differential regulation of downstream signaling molecules. Collectively, our studies show that different spatiotemporal dynamics of PIP3 and PI34P2 and their ability to selectively activate key signaling proteins allow them to mediate class I PI3K signaling pathways in a spatiotemporally specific manner.


Subject(s)
Optical Imaging/methods , Phosphatidylinositol Phosphates/physiology , Single Molecule Imaging/methods , Animals , Cell Line , Cell Membrane , Humans , Inositol Phosphates , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositols , Protein Isoforms , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...