Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Med Sci Monit ; 26: e922598, 2020 Aug 09.
Article in English | MEDLINE | ID: mdl-32772042

ABSTRACT

BACKGROUND For proximal humeral fractures (PHFs), locking intramedullary nails and locking plates have been widely used. However, few reports have been published on the therapy of complex PHFs accompanying humeral shaft fractures. Therefore, we performed this research to analyze the effectiveness of locking intramedullary nails and locking plates in the management of proximal humeral fractures involving the humeral shaft. MATERIAL AND METHODS We retrospectively reviewed 40 cases diagnosed with proximal humeral fractures involving the humeral shaft fixed with either locking intramedullary nails or locking plates with at least of 2 years' follow-up. Clinical data were obtained from the medical records. Follow-up data included the Constant-Murley score, American Shoulder and Elbow Surgeons score (ASES), visual analog scale score (VAS), and the relative strength of the supraspinatus and deltoid muscles. RESULTS In total, 19 locking plate patients and 21 locking intramedullary nail patients were analyzed. The average follow-up period was 35 months in the locking plate group and 34 months in the locking intramedullary nail group. There were obvious differences in the intraoperative blood loss, time of operation, and the length of operative incision between the 2 groups (p<0.05). There were no significant differences between the groups in Constant-Murley score, ASES, VAS, or the relative strength of supraspinatus and deltoid muscles. CONCLUSIONS For PHFs involving the humeral shaft, both locking plates and locking intramedullary nails can achieve satisfactory functional results in the long-term follow-up assessment. The locking intramedullary nail group was superior with regards to intraoperative blood loss, time of operation, and length of incision.


Subject(s)
Bone Nails , Fracture Fixation, Intramedullary/methods , Shoulder Fractures/surgery , Female , Humans , Male , Middle Aged , Postoperative Complications/physiopathology , Retrospective Studies , Shoulder Fractures/rehabilitation
2.
J Exp Clin Cancer Res ; 42(1): 136, 2023 May 31.
Article in English | MEDLINE | ID: mdl-37254126

ABSTRACT

BACKGROUND: Increasing evidence indicates that the tumor microenvironment (TME) is a crucial determinant of cancer progression. However, the clinical and pathobiological significance of stromal signatures in the TME, as a complex dynamic entity, is still unclear in esophageal squamous cell carcinoma (ESCC). METHODS: Herein, we used single-cell transcriptome sequencing data, imaging mass cytometry (IMC) and multiplex immunofluorescence staining to characterize the stromal signatures in ESCC and evaluate their prognostic values in this aggressive disease. An automated quantitative pathology imaging system determined the locations of the lamina propria, stroma, and invasive front. Subsequently, IMC spatial analyses further uncovered spatial interaction and distribution. Additionally, bioinformatics analysis was performed to explore the TME remodeling mechanism in ESCC. To define a new molecular prognostic model, we calculated the risk score of each patient based on their TME signatures and pTNM stages. RESULTS: We demonstrate that the presence of fibroblasts at the tumor invasive front was associated with the invasive depth and poor prognosis. Furthermore, the amount of α-smooth muscle actin (α-SMA)+ fibroblasts at the tumor invasive front positively correlated with the number of macrophages (MØs), but negatively correlated with that of tumor-infiltrating granzyme B+ immune cells, and CD4+ and CD8+ T cells. Spatial analyses uncovered a significant spatial interaction between α-SMA+ fibroblasts and CD163+ MØs in the TME, which resulted in spatially exclusive interactions to anti-tumor immune cells. We further validated the laminin and collagen signaling network contributions to TME remodeling. Moreover, compared with pTNM staging, a molecular prognostic model, based on expression of α-SMA+ fibroblasts at the invasive front, and CD163+ MØs, showed higher accuracy in predicting survival or recurrence in ESCC patients. Regression analysis confirmed this model is an independent predictor for survival, which also identifies a high-risk group of ESCC patients that can benefit from adjuvant therapy. CONCLUSIONS: Our newly defined biomarker signature may serve as a complement for current clinical risk stratification approaches and provide potential therapeutic targets for reversing the fibroblast-mediated immunosuppressive microenvironment.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Carcinoma, Squamous Cell/pathology , CD8-Positive T-Lymphocytes/metabolism , Prognosis , Fibroblasts/metabolism , Tumor Microenvironment
3.
Microsurgery ; 30(4): 314-20, 2010 May.
Article in English | MEDLINE | ID: mdl-20058301

ABSTRACT

The purpose of our study was to establish the profile of cortical reorganization in whole BPAI on rats and evaluate changes of cortical reorganization after repair of the median nerve with the contralateral C7 root transfer. Forty adult SD rats underwent whole roots avulsion of left brachial plexus, among them 20 received contralateral C7 root transfer to the injured median nerve. Intracortical microstimulation was performed in primary motor cortex (M1) at intervals of 3, 5, 7, and 10 months, postoperatively. The maps of motor cortical responses were constructed. Five normal rats were used as the control. Results showed that stimulating right M1 elicited motion of left vibrissae, submaxilla, neck, back, and left hindlimb after left BPAI, among them neck representation area replaced the forelimb area throughout the reorganization process. The left forelimb representation area was found in the left motor cortex 5 months after the contralateral C7 root transfer and existed in both motor cortexes at 7th postoperative month. The left forelimb representation area was detected only in right motor cortex at 10th month, postoperatively. In conclusions, after the contralateral C7 root transfer for repair of the median nerve in BPAI, the cortical reorganization occurred in a time-dependent reorganization. The findings from this study demonstrate that brain involves in the functional recovery after BPAI and repair with nerve transfer and suggest that efforts to improve the results from nerve repair should address the peripheral nerve as well as the brain.


Subject(s)
Brachial Plexus Neuropathies/surgery , Motor Cortex/physiopathology , Nerve Regeneration/physiology , Nerve Transfer/methods , Spinal Nerve Roots/surgery , Anastomosis, Surgical , Animals , Brachial Plexus/injuries , Brachial Plexus/surgery , Brain Mapping/methods , Cervical Vertebrae , Disease Models, Animal , Evoked Potentials, Motor , Female , Male , Median Nerve/surgery , Neural Conduction/physiology , Random Allocation , Rats , Rats, Sprague-Dawley , Recovery of Function , Reference Values
4.
Zhonghua Bing Li Xue Za Zhi ; 37(5): 328-32, 2008 May.
Article in Zh | MEDLINE | ID: mdl-18956652

ABSTRACT

OBJECTIVE: To investigate the role of focal adhesion kinase (FAK) in cardiac hypertrophy induced by hypertension. METHODS: Using immunofluorescent labeling, confocal microscopy and Western blot, the expression and subcellular location of FAK-pSer722 and FAK-pSer910 were determined in cardiac myocytes of the left ventricles from 2, 6, 12, and 18 month-old spontaneously hypertensive heart failure (SHHF) rats and age-matched Wistar-Kyoto (WKY) control rats, respectively. RESULTS: There was no obvious difference in FAK-pSer722 and FAK-pSer910 expression between 2 month-old SHHF and WKY rats. In contrast with the control groups, the expression of FAK-pSer722 and FAK-pSer910 significantly increased in cardiac myocytes of the left ventricle, from 6, 12 and 18 month-old SHHF rats. Both FAK-pSer722 and FAK-pSer910 were translocated and acummulated in nuclei of cardiac myocytes from 6, 12, and 18 month-old SHHF rats. CONCLUSION: Phosphorylation and translocation of serine 722 and serine 910 of phosphorylated FAK play an important role in the de-compensatory cardiac hypertrophy.


Subject(s)
Cardiomegaly/enzymology , Cell Nucleus/enzymology , Focal Adhesion Kinase 1/metabolism , Heart Ventricles/pathology , Hypertension/enzymology , Myocytes, Cardiac/enzymology , Serine/metabolism , Signal Transduction/drug effects , Animals , Cardiomegaly/metabolism , Cell Nucleus/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/physiology , Heart Failure , Hypertrophy/enzymology , Myocytes, Cardiac/pathology , Phosphorylation , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction/physiology
5.
Zhonghua Bing Li Xue Za Zhi ; 36(10): 677-80, 2007 Oct.
Article in Zh | MEDLINE | ID: mdl-18194601

ABSTRACT

OBJECTIVES: To investigate the role of focal adhesion kinase (FAK) in the pathogenesis of cardiac hypertrophy induced by hypertension. METHODS: Using immunofluorescent labeling, confocal microscopy and Western blotting, the expression and subcellular localization of FAK in the cardiac myocytes of left ventricle were determined in 2, 6, 12, and 18 month-old rats with spontaneously hypertensive heart failure (SHHF) along with age-matched control Wistar-Kyoto (WKY) rats. RESULTS: There was no significant difference of FAK expression between 2 month-old SHHF and WKY rats (50.5+/-6.9 vs. 49.8+/-5.0, n=6, P>0.05). In contrast with the control groups, the expression of FAK significantly increased in 6, 12 and 18 month-old SHHF rats (130.6+/-3.0 vs. 47.3+/-1.3, 144.7+/-5.4 vs. 46.4+/-3.1, 141.4+/-9.8 vs. 48.5+/-2.2, each groups n=6, P<0.05) with FAK protein primarily cumulated in the intercalated disks and nuclei. CONCLUSIONS: FAK may play a role in the cell signaling transduction leading to cardiac hypertrophy, presumably through regulations of hypertrophic gene transcription and RNA processing.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Heart Ventricles/pathology , Hypertension/complications , Hypertrophy, Left Ventricular/enzymology , Myocytes, Cardiac/metabolism , Animals , Hypertrophy, Left Ventricular/etiology , Male , Microscopy, Confocal , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction
6.
Brain Res ; 1471: 93-101, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22771398

ABSTRACT

It has been demonstrated that there could be long range interhemispheric reorganization between bilateral hemispheres after peripheral cross nerve transfer. Our previous studies found a striking dynamic process of interhemispheric functional reorganization in adult rats with cross seventh cervical nerve transfer. But it remains a question whether the extensive interhemispheric functional reorganization after cross nerve transfer depends on connectivities at the cortical or subcortical level. In the present study, 18 rats with cross C7 transfer were concurrently treated with corpus callosotomy while the other 18 were not. Intracortical microstimulation was performed in the primary motor cortex (M1) at intervals of 5, 7, and 10 months postoperatively. The neural electrophysiology study showed that the representation of the injured forepaw appeared in the ipsilateral cortex at 5 months after the cross nerve transfer combined with corpus callosotomy, and it shared great overlapping zones with the representation of the health forepaw. And then, at 7-10 months, the cortical representation of the paralyzed forepaw was still located in the ipsilateral motor cortex, although significantly contracted. In contrast, rats with mere cross nerve transfer still presented interhemispheric reorganization. The results indicated that corpus callosotomy in the early stage after cross C7 transfer may had interrupted the interhemispheric functional reorganization. Combined the present study with our previous research findings, we explored the possible pathway and mechanisms of the interhemispheric functional reorganization. Thus we came to the conclusion that interhemispheric connectivity at the cortical level was essential in establishing the new contralateral control of the paralyzed limb at the initial stage after cross nerve transfer.


Subject(s)
Functional Laterality/physiology , Motor Cortex/physiology , Nerve Transfer/methods , Neural Pathways/physiology , Radiculopathy , Animals , Brain Mapping , Corpus Callosum/injuries , Corpus Callosum/physiology , Disease Models, Animal , Forelimb/innervation , Hand Strength/physiology , Median Nerve/physiology , Median Nerve/transplantation , Radiculopathy/pathology , Radiculopathy/physiopathology , Radiculopathy/surgery , Rats , Rats, Sprague-Dawley , Time Factors
7.
Neurosurgery ; 67(1): 136-43; discussion 143, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20559101

ABSTRACT

BACKGROUND: The restoration of sensory and motor function in brachial plexus root avulsion patients is a difficult challenge. The central nervous system plays an important role in sensory recovery after peripheral nerve injury and repair. OBJECTIVE: To investigate the sensory restoration process after surgery at the cortical level in rodent models with a contralateral C7 nerve transfer. METHODS: Thirty-five male Sprague-Dawley rats were used in this experiment, and both behavioral tests and somatosensory evoked potentials were used to investigate the sensory function recovery of the injured forepaws and the cortical reorganization in the rats postoperatively. RESULTS: The results demonstrated a dynamic change in the ipsilateral somatosensory cortex, both in the shape and location, where overlapping sensory cortical representations of the healthy and injured forepaws were observed consistently. Behavioral tests show that the sensation first occurred only in the healthy forepaw and later in both when stimulating the injured one, which suggested a tendency of the sensation function to recover in the injured forepaws of the rats as time progressed. CONCLUSION: The cortical reorganization occurred only in the ipsilateral hemisphere, which is different from the motor cortex reorganization using the same model as that described in a previous study. This reorganization pattern offers an interpretation of the unique sensory recovery process after the transfer of the C7 nerve to the contralateral median nerve, but also provides the basis for further sensory restoration in clinical practice.


Subject(s)
Brachial Plexus Neuropathies/physiopathology , Brachial Plexus Neuropathies/surgery , Forelimb/innervation , Nerve Transfer/methods , Recovery of Function/physiology , Sensation Disorders/physiopathology , Sensation Disorders/surgery , Animals , Brachial Plexus Neuropathies/complications , Disease Models, Animal , Forelimb/injuries , Forelimb/physiopathology , Male , Median Nerve/physiology , Median Nerve/surgery , Rats , Rats, Sprague-Dawley , Sensation Disorders/etiology
SELECTION OF CITATIONS
SEARCH DETAIL